Genmab Announces Positive Topline Results in Phase III ANDROMEDA Study of Daratumumab in Light-chain (AL) Amyloidosis – GlobeNewswire

Company Announcement

Copenhagen, Denmark; May 28, 2020 Genmab A/S (Nasdaq: GMAB) announced today positive topline results from the Phase III ANDROMEDA (AMY3001) study of subcutaneous (SC) daratumumab in combination with cyclophosphamide, bortezomib and dexamethasone (CyBorD) for patients with newly diagnosed light-chain (AL) amyloidosis. The study, conducted by Janssen Biotech, Inc. (Janssen), met the primary endpoint of percentage of patients with hematologic complete response. Patients in the study treated with daratumumab in combination with CyBorD had a 53.3% hematologic complete response compared to 18.1% of patients who were treated with CyBorD alone (odds ratio of 5.1 (95% CI 3.2 8.2, p<0.0001)).

Overall, the safety profile of daratumumab SC in combination with CyBorD is consistent with the known safety profile of the CyBorD regimen and the known safety profile of daratumumab.

We are very pleased with the topline results from the Phase III ANDROMEDA study in AL amyloidosis. We believe the data supports the potential of daratumumab in the treatment of this devastating, progressive disease, for which no approved treatments are available, said Jan van de Winkel, Ph.D., Chief Executive Officer of Genmab.

Janssen, which obtained an exclusive worldwide license to develop, manufacture and commercialize daratumumab from Genmab in 2012, will discuss with health authorities the potential for a regulatory submission for this indication.

About the ANDROMEDA (AMY3001) studyThe Phase III study (NCT03201965) included 388 patients newly diagnosed with AL amyloidosis. Patients were randomized to receive treatment with either subcutaneous daratumumab in combination with cyclophosphamide (a chemotherapy), bortezomib (a proteasome inhibitor) and dexamethasone (a corticosteroid) or treatment with cyclophosphamide, bortezomib and dexamethasone alone. The primary endpoint of the study is the percentage of patients who achieve hematologic complete response.

About Light-chain (AL) AmyloidosisAmyloidosis is a disease that occurs when amyloid proteins, which are abnormal proteins, accumulate in tissues and organs. When the amyloid proteins cluster together, they form deposits that damage the tissues and organs. AL amyloidosis most frequently affects the heart, kidneys, liver, nervous system and digestive tract. There is currently no cure or existing approved therapies for AL amyloidosis though it can be treated with chemotherapy, dexamethasone, stem cell transplants and supportive therapies.1 It is estimated that there are approximately 3,000 to 4,000 new cases of AL amyloidosis diagnosed annually in the U.S.2

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.3 DARZALEX is the first monoclonal antibody (mAb) to receive U.S. Food and Drug Administration (U.S. FDA) approval to treat multiple myeloma. DARZALEX intravenous infusion is indicated for the treatment of adult patients in Europe: 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 therapy4. 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.5 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).3,5,6,7,8,9

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.

Contact: Marisol Peron, Corporate Vice President, Communications & Investor Relations T: +1 609 524 0065; E: mmp@genmab.com

For Investor Relations: Andrew Carlsen, Senior Director, Investor RelationsT: +45 3377 9558; E: acn@genmab.com

This Company Announcement contains forward looking statements. The words believe, expect, anticipate, intend and plan and similar expressions identify forward looking statements. Actual results or performance may differ materially from any future results or performance expressed or implied by such statements. The important factors that could cause our actual results or performance to differ materially include, among others, risks associated with pre-clinical and clinical development of products, uncertainties related to the outcome and conduct of clinical trials including unforeseen safety issues, uncertainties related to product manufacturing, the lack of market acceptance of our products, our inability to manage growth, the competitive environment in relation to our business area and markets, our inability to attract and retain suitably qualified personnel, the unenforceability or lack of protection of our patents and proprietary rights, our relationships with affiliated entities, changes and developments in technology which may render our products or technologies obsolete, and other factors. For a further discussion of these risks, please refer to the risk management sections in Genmabs most recent financial reports, which are available on http://www.genmab.com and the risk factors included in Genmabs most recent Annual Report on Form 20-F and other filings with the U.S. Securities and Exchange Commission (SEC), which are available at http://www.sec.gov. Genmab does not undertake any obligation to update or revise forward looking statements in this Company Announcement nor to confirm such statements to reflect subsequent events or circumstances after the date made or in relation to actual results, unless required by law.

Genmab A/S and/or its subsidiaries own the following trademarks: Genmab; the Y-shaped Genmab logo; Genmab in combination with the Y-shaped Genmab logo; HuMax; DuoBody; DuoBody in combination with the DuoBody logo; HexaBody; HexaBody in combination with the HexaBody logo; DuoHexaBody; HexElect; and UniBody. Arzerra is a trademark of Novartis AG or its affiliates. DARZALEX and DARZALEX FASPRO are trademarks of Janssen Pharmaceutica NV. TEPEZZA is a trademark of Horizon Therapeutics plc.

1 Mayo Clinic website: http://www.mayoclinic.com/health/amyloidosis/DS004312 Research and Markets, Amyloidosis Treatment Market Size, Share & Trends Analysis Report by Treatment (Stem Cell Transplant, Chemotherapy, Supportive Care, Surgery, Targeted Therapy), By Country, And Segment Forecasts, 2018 - 20253 DARZALEX Prescribing information, April 2020. Available at: https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/761036s027lbl.pdf Last accessed April 20204 DARZALEX Summary of Product Characteristics, available at https://www.ema.europa.eu/en/medicines/human/EPAR/darzalex Last accessed October 20195 DARZALEX FASPRO Prescribing information, May 2020. Available at: https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/761145s000lbl.pdf Last accessed May 20206 De Weers, M et al. Daratumumab, a Novel Therapeutic Human CD38 Monoclonal Antibody, Induces Killing of Multiple Myeloma and Other Hematological Tumors. The Journal of Immunology. 2011; 186: 1840-1848.7 Overdijk, MB, et al. Antibody-mediated phagocytosis contributes to the anti-tumor activity of the therapeutic antibody daratumumab in lymphoma and multiple myeloma. MAbs. 2015; 7: 311-21.8 Krejcik, MD et al. Daratumumab Depletes CD38+ Immune-regulatory Cells, Promotes T-cell Expansion, and Skews T-cell Repertoire in Multiple Myeloma. Blood. 2016; 128: 384-94.9Jansen, JH et al. Daratumumab, a human CD38 antibody induces apoptosis of myeloma tumor cells via Fc receptor-mediated crosslinking.Blood. 2012; 120(21): abstract 2974

Company Announcement no. 22CVR no. 2102 3884LEI Code 529900MTJPDPE4MHJ122

Genmab A/SKalvebod Brygge 431560 Copenhagen VDenmark

Read the original post:
Genmab Announces Positive Topline Results in Phase III ANDROMEDA Study of Daratumumab in Light-chain (AL) Amyloidosis - GlobeNewswire

Myeloma cells shift osteoblastogenesis to adipogenesis by inhibiting the ubiquitin ligase MURF1 in mesenchymal stem cells – Science

How myeloma promotes bone loss

Multiple myeloma can lead to bone loss by reducing the differentiation of mesenchymal stem cells (MSCs) into osteoblasts. Using a combination of single-cell RNA sequencing, in vitro coculture, and experiments with human myeloma cells and MSCs in mice, Liu et al. demonstrated how direct contact between myeloma cells and MSCs shifted the balance of MSC differentiation to favor adipogenesis over osteoblastogenesis. Integrin 4 on the surface of myeloma cells activated the adhesion molecule VCAM1 on MSCs, leading to protein kinase C 1 (PKC1)dependent repression of the E3 ubiquitin ligase MURF1 and subsequent stabilization of the adipocyte transcription factor PPAR2. These findings suggest a possible avenue for preventing or treating myeloma-induced bone loss in patients.

The suppression of bone formation is a hallmark of multiple myeloma. Myeloma cells inhibit osteoblastogenesis from mesenchymal stem cells (MSCs), which can also differentiate into adipocytes. We investigated myeloma-MSC interactions and the effects of such interactions on the differentiation of MSCs into adipocytes or osteoblasts using single-cell RNA sequencing, in vitro coculture, and subcutaneous injection of MSCs and myeloma cells into mice. Our results revealed that the 4 integrin subunit on myeloma cells stimulated vascular cell adhesion molecule1 (VCAM1) on MSCs, leading to the activation of protein kinase C 1 (PKC1) signaling and repression of the muscle ring-finger protein-1 (MURF1)mediated ubiquitylation of peroxisome proliferatoractivated receptor 2 (PPAR2). Stabilized PPAR2 proteins enhanced adipogenesis and consequently reduced osteoblastogenesis from MSCs, thus suppressing bone formation in vitro and in vivo. These findings reveal that suppressed bone formation is a direct consequence of myeloma-MSC contact that promotes the differentiation of MSCs into adipocytes at the expense of osteoblasts. Thus, this study provides a potential strategy for treating bone resorption in patients with myeloma by counteracting tumor-MSC interactions.

More than 80% of patients with multiple myeloma suffer from bone destruction, which greatly reduces their quality of life and has a severe negative impact on survival (1). New bone formation, which usually occurs at sites of previously resorbed bone, is strongly suppressed in patients with myeloma, and bone destruction rarely heals in these patients (2). Therefore, prevention of bone disease is a priority in myeloma treatment, and understanding the mechanisms by which myeloma cells disturb the bone marrow (BM) is fundamental to myeloma-associated bone diseases.

Osteoblasts originate from mesenchymal stem cells (MSCs) and are responsible for bone formation. It has been reported that myeloma cells inhibit MSC differentiation into mature osteoblasts (35). Osteoblasts and adipocytes arise from a common MSC-derived progenitor and exhibit lineage plasticity, which further complicates the relationship between these two cell types in myeloma cellinfiltrated BM (6). Traditionally, initiation of adipogenesis and osteogenesis has been widely regarded as mutually exclusive, and factors that inhibit osteoblastogenesis activate adipogenesis and vice versa (7). Previous studies have demonstrated that MSCs differentiate into either adipocytes or osteoblasts depending on the stimulator (8), and adipocytes transdifferentiate into osteoblasts in patients with several benign diseases (9). However, the underlying effects of myeloma cells on the activation of adipogenic transcriptional factors and the molecular mechanisms involved are still obscure.

Peroxisome proliferatoractivated receptor 2 (PPAR2) is a key transcription factor for the regulation of fatty acid storage and glucose metabolism (10), and it activates genes important for adipocyte differentiation and function (11). Previous findings have demonstrated that PPAR2 plays important roles not only in the activation of adipogenesis but also in the suppression of osteoblastogenesis (12, 13). In vitro coculture of MSCs from multiple myeloma patients with malignant plasma cell lines enhances adipocyte differentiation of the MSCs due to increased PPAR2 in the MSCs (14), suggesting that PPAR2 mediates myeloma-induced adipogenesis. However, the mechanism by which myeloma cells activate PPAR2 in MSCs, thereby causing MSCs to differentiate into adipocytes rather than osteoblasts, remains unclear.

In the present study, we demonstrated that myeloma cells enhanced the differentiation of human MSCs into adipocytes rather than osteoblasts by stabilizing PPAR2 protein through an integrin 4protein kinase C 1 (PKC1)muscle ring-finger protein-1 (MURF1) signaling pathway in MSCs. Our study thus provides a potential therapeutic strategy for myeloma-associated bone disease.

To determine whether myeloma cells affect MSC fate, we characterized the heterogeneity of human BMderived MSCs after exposure to myeloma cells. We cultured MSCs alone (controls) or cocultured them with myeloma cells in a 1:1 mixture of adipocyte:osteoblast (1:1 AD:OB) medium (Fig. 1A). An aliquot of cells was cultured for 48 hours and then subjected to single-cell RNA sequencing (scRNA-seq). We cultured another aliquot of cells for 2 weeks, removed the myeloma cells, and assessed the ability of the MSCs to differentiate into mature osteoblasts or adipocytes using Alizarin red-S, which stains calcium deposits, and Oil red O, which stains lipids (Fig. 1A). Trajectory analysis indicated the dynamic cellular transition processes of MSCs in vitro, in line with the in vivo MSC fates, reported by Wolock et al. (15). We observed a fate shift in MSC differentiation when MSCs were cocultured with myeloma cells (Fig. 1B). T-distributed stochastic neighbor embedding cluster analysis based on the entire transcriptome gene signature showed that both control and cocultured MSCs had specific transcriptome characteristics (Fig. 1C). After identification of genes with highly variable expression across the dataset, clusters were identified in each of the control and coculture groups (Fig. 1C). Enrichment analysis demonstrated that the adipokine signaling pathway and the mineral absorption pathway were among the 20 pathways most significantly changed in MSCs cocultured with myeloma cells (Fig. 1D). We identified clusters 0, 1, 6, and 8 in the MSCs cocultured with myeloma cells as being of adipogenic lineage because their expression of the specific markers of adipogenesis, the ADD1 and PPAR genes, were markedly higher than that of other clusters (Fig. 1E). These results demonstrated that myeloma cells at least partially increase MSC transformation into adipocytes.

(A) System for coculturing of human MSCs with the human multiple myeloma cell (MM) line MM.1S in a 1:1 mixture of adipocyte (AD) and osteoblast (OB) medium. Cells were cocultured for 48 hours and then MSC-derived cells were subjected to single-cell RNA sequencing (scRNA-seq). As a control, scRNA-seq was also performed on MSCs cultured alone in 1:1 AD:OB medium. (B) The single-cell trajectory reconstructed by Monocle in the control (Ctrl) and coculture (Coculture) groups. Each point represents a cell, and colors indicate their respective group. n = 2 independent experiments. The trajectory constructed by Monocle is in black. (C) T-distributed stochastic neighbor embedding (t-SNE) plot depicting clusters of MSCs cultured alone (Ctrl) or cocultured with MM cells. The first two dimensions are shown. Each cluster represents individual cells with similar transcriptional profiles of MSCs or different MSC lineages, with total of 10 clusters from aggregated samples of two biologically independent experiments. (D) Enrichment analysis showing the 20 most significantly changed pathways in the MSCs cocultured with MM cells. Red indicates activated pathways, and green indicates repressed pathways. (E) Distributions of unique transcripts per cell and PPARG and CEBPB gene expression in all cell clusters. The red frame shows the highest expression among the clusters. TGF-, transforming growth factor.

The coculture of MSCs and myeloma cells resulted in lower Alizarin red-S staining and higher Oil red O staining in MSCs, indicating an increase in the generation of adipocytes, compared to culture of MSCs alone (Fig. 2A). We further labeled cocultured MSCs with antibodies recognizing the osteoblast marker osteocalcin or the adipocyte marker fatty acid binding protein 4 (FABP4) and analyzed them using flow cytometry. We observed that culturing MSCs in osteoblast medium increased the osteocalcin+ population and that coculturing MSCs with myeloma cells inhibited this increase. Also, culturing MSCs in adipocyte medium increased the FABP4+ population, and coculturing them with myeloma cells further increased it. When we cultured MSCs alone in the 1:1 AD:OB medium, both the osteocalcin+ and FABP4+ populations increased, whereas coculturing MSCs with myeloma cells reduced the osteocalcin+ population but increased the FABP4+ population (Fig. 2, B and C). We obtained similar effects on osteoblastogenesis (Fig. 2D) and adipogenesis (Fig. 2E) when we cocultured MSCs with six other myeloma cell lines or with CD138+ primary myeloma cells isolated from BM aspirates from five patients with myeloma, but not with plasma cells from healthy donors (Fig. 2, F and G). Real-time polymerase chain reaction (PCR) analysis further showed lower expression of the osteoblast differentiationassociated genes alkaline phosphatase (ALP), secreted phosphoprotein 1 (SPP1), collagen type I alpha 1 chain (COL1A1), and bone gamma-carboxyglutamate protein (BGLAP; Fig. 2H) and higher expression of the adipocyte differentiationassociated genes delta-like noncanonical Notch ligand 1 (DLK1), diacylglycerol O-acyltransferase 1 (DGAT1), FABP4, and fatty acid synthase (FASN; Fig. 2I) in MSCs cocultured with ARP-1 or MM.1S myeloma cells than in MSCs cultured alone. These results demonstrate that myeloma cells directed the differentiation of MSCs preferentially toward adipocytes than to osteoblasts.

(A) Representative images of Alizarin red-S and Oil red O staining (whole wells and enlarged views) of MSCs cultured alone or cocultured with ARP-1 or MM.1S myeloma cell lines in MSC medium, adipocyte (AD) medium, osteoblast (OB) medium, or mixed 1:1 AD:OB medium as indicated. n = 3 independent experiments. Scale bars, 5 mm (whole wells) and 20 m (enlargements). (B and C) Flow cytometric analysis showing the percentage of osteocalcin+ (B) and FABP4+ (C) cells in cultures of MSCs alone or in direct contact with ARP-1 cells in the indicated medium. Data are representative of three independent experiments with each sample analyzed in triplicate. (D and E) Quantification of Alizarin red-S (D) and Oil red O (E) staining of MSCs cultured alone (No MM) or cocultured with the six indicated myeloma cell lines. Combined data are from three biologically independent experiments. (F and G) Quantification of Alizarin red-S (F) and Oil red O (G) staining of MSCs cultured alone or cocultured with primary myeloma cells isolated from BM aspirates of five patients with myeloma (P1 to P5) or normal plasma cells from the BM of two healthy donors (PC1 and PC2). Combined data are from n = 3 experiments using the same donor source material. (H and I) Quantitative reverse transcription PCR showing the expression of the osteoblast differentiationassociated genes ALP, SPP1, COL1A1, and BGLAP (H) and the adipocyte differentiationassociated genes DLK1, DGAT1, FABP4, and FASN (I) in cells generated by coculture of MSCs with myeloma cells relative to expression of each gene in MSCs cultured alone. Combined data are from n = 3 independent experiments. All data are means SD. *P 0.05 and **P 0.01. P values were determined using one-way ANOVA with Tukeys multiple comparisons test.

We next investigated the mechanism of myeloma-induced shifting of MSCs from osteoblastogenesis to adipogenesis. We focused on PPAR2 because it is a key transcriptional factor for the activation of adipogenesis. scRNA-seq showed higher PPAR2 mRNA expression in MSCs cocultured with myeloma cells compared to MSCs cultured alone (Fig. 1E). Using the coculture system with MSCs and myeloma cells in a 1:1 mixture of adipocyte and osteoblast medium, we again observed the transformation of osteoblastogenesis into adipogenesis in MSCs cocultured with myeloma cells (Fig. 3A), as well as an increase in the abundance of PPAR2 in MSCs cultured with myeloma cells (Fig. 3B and fig. S1). To determine the importance of PPAR2 in MSC transformation, we added the PPAR2 antagonist G3335 to cocultures. G3335 inhibited the myeloma cellinduced increase in PPAR2 protein (Fig. 3B and fig. S1). Consistent with the Western blot results, G3335 treatment decreased Oil red O staining (Fig. 3C) and adipocyte gene expression (Fig. 3D) and increased Alizarin red-S staining (Fig. 3E) and osteoblast gene expression (Fig. 3F). These results suggest that PPAR2 mediated myeloma-induced MSC transformation into adipocytes.

(A) Representative images of Oil red O or Alizarin red-S staining of MSCs cultured alone or cocultured with ARP-1 or MM.1S myeloma cells in 1:1 OB:AD medium and treated with the PPAR2 antagonist G3335 as indicated. Scale bar, 5 mm. (B) Representative Western blot for PPAR2 in cells treated as in (A). Quantitation is presented in fig. S1. Actin is a loading control. (C to F) Quantitative analysis of Oil red O staining (C), adipocyte differentiationassociated gene expression (D), Alizarin red-S staining (E), and osteoblast differentiationassociated gene expression (F) in cells treated as in (A). Data are means SD from n = 3 independent experiments. *P 0.05 and **P 0.01. P values were determined using Students t test for paired samples (D and F) and one-way ANOVA with Tukeys multiple comparisons test (C and E).

To determine whether myeloma cells distort MSC transformation through myeloma-secreted soluble factors or cell-to-cell contact, we cocultured MSCs with ARP-1 or MM.1S myeloma cells in 1:1 AD:OB medium either together or separated by transwell inserts. We observed that the transwell coculture had a slight effect on increased Oil red O staining, whereas cell-to-cell contact coculture in the mixed medium produced much more significant boost of this staining, suggesting that direct interaction between MSCs and myeloma cells was needed for enhancing adipogenesis from MSCs (Fig. 4A). When we added supernatants collected from 24-hour cultures of ARP-1 or MM.1S cells to MSC cultures, we obtained results similar to those for the transwell coculture (Fig. 4A), reaffirming the importance of direct contact of MSCs with myeloma cells.

(A) Oil red O staining in MSCs cultured alone (No MM) or cocultured with ARP-1 or MM.1S myeloma cells in 1:1 AD:OB medium directly (cell-cell) or separated by transwell inserts (Trans) or in myeloma cell culture media (sup). Staining was quantified relative to the No MM condition. Representative data are from three independent experiments. (B to D) Relative Oil red O staining (B) and the relative expression of the indicated osteoblast (C) and adipocyte (D) marker genes in MSCs cultured alone (No MM) or cocultured with ARP-1 or MM.1S cells with or without neutralizing antibodies against integrin subunits 4, 5, V, or L. Combined data are from three independent experiments. (E) Western blot showing integrin 4 and integrin 1 in ARP-1 and MM.1S cells expressing shRNA targeting integrin 4 (4 KD) or nontargeted control shRNAs (NT Ctrl). Actin is a loading control. (Blot is a representative of three independent experiments, and blot quantitation data are presented in fig. S2C. (F to J) PPAR2 protein (F), Alizarin red-S staining (G), Oil red O staining (H), osteoblast marker gene expression (I), and adipocyte marker gene expression (J) in MSCs cultured alone or cocultured with ARP-1 or MM.1S cells expressing NT Ctrl or 4 KD shRNA. Blots in (E) and (F) are representative of three independent experiments, and blot quantitation is presented in fig. S2 (A and D). Data in (G) to (J) are means SD from n = 3 independent experiments using MSCs derived from BM aspirates of three healthy donors. Data are **P 0.01. P values were determined using one-way ANOVA with Tukeys multiple comparisons test.

To identify the specific molecules involved in adipocyte differentiation, we tested the effect of blocking antibodies against various integrins, which are highly expressed in myeloma cells, in cocultures of MSCs with ARP-1 or MM.1S cells in 1:1 AD:OB medium. The addition of an antibody against integrin 4but not antibodies against integrins 5, V, or L or a control immunoglobulin G (IgG)markedly reduced Oil red O staining in cocultures with both myeloma cell lines (Fig. 4B). The addition of the antibody recognizing integrin 4 to cocultures of MSCs and ARP-1 cells in the mixed medium also increased osteoblast gene expression (Fig. 4C) and decreased adipocyte gene expression (Fig. 4D) substantially more than did the addition of the control IgG. To determine whether integrin 4 affected PPAR2 production in MSCs, we infected ARP-1 and MM.1S cells with a lentivirus carrying short hairpin RNAs (shRNAs) targeting integrin 4 (fig. S2A). Integrin 4 knockdown (4 KD) reduced integrin 4 production without changing the cell viability or proliferation, whereas integrin 1 remained unchanged in ARP-1 and MM.1S cells (Fig. 4E and fig. S2, A to C). We also cocultured MSCs with control or 4 KD myeloma cells in the mixed medium. Western blot analysis demonstrated that 4 KD in myeloma cells reduced PPAR2 protein production in MSCs more than did myeloma cells expressing a nontargeting control shRNA (Fig. 4F and fig. S2D). In addition, coculture of MSCs with 4 KD myeloma cells induced higher Alizarin red-S staining (Fig. 4G) and osteoblast gene expression (Fig. 4H) but lower Oil red O staining (Fig. 4I) and adipocyte gene expression (Fig. 4J) compared to MSCs cocultured with myeloma cells expressing the control shRNA.

Because vascular cell adhesion molecule1 (VCAM1) is a major ligand of integrin 4, we investigated whether it mediated myeloma-induced MSC transformation by adding a blocking antibody against VCAM1 or control IgG to MSC and myeloma cell cocultures. Addition of the antibody, but not IgG, increased Alizarin red-S staining (Fig. 5A) and osteoblast gene expression (Fig. 5B) but decreased Oil red O staining (Fig. 5C) and adipocyte gene expression (Fig. 5D) in MSCs. To determine whether binding of integrin 4 to VCAM1 induced an increase in PPAR2, we constructed MSCs with reduced expression of VCAM1 using a lentivirus carrying VCAM1 shRNAs (VCAM1 KD) (Fig. 5E and fig. S3A) and cocultured myeloma cells with control or VCAM1 KD MSCs. Western blot analysis showed that cocultured VCAM1 KD MSCs had reduced PPAR2 protein production compared to cocultured MSCs expressing nontargeting control shRNA (Fig. 5F and fig. S3B). We also found that VCAM1 KD in MSCs considerably abrogated myeloma-induced suppression of osteoblastogenesis and activation of adipogenesis, because Oil red O staining and adipocyte gene expression decreased significantly (Fig. 5, G and H), whereas Alizarin red-S staining and osteoblast gene expression both increased (Fig. 5, I and J).

(A to D) Alizarin red-S staining (A), Oil red O staining (B), and real-time PCR analysis of the expression of osteoblast (C) and adipocyte (D) marker genes in MSCs cultured alone (No MM) or cocultured with ARP-1 or MM.1S myeloma cells in the presence of a neutralizing antibody against VCAM1 or IgG (control). Data are from n = 3 independent experiments. (E) Western blotting analysis showing VCAM1 in the MSCs infected with a lentivirus carrying nontargeted control shRNAs (NT Ctrl-MSCs) or human VCAM1 shRNAs (VCAM1 KD-MSCs). Actin is a loading control. Blot is a representative of three independent experiments, and blot quantitation is presented in fig. S3A. (F to J) PPAR2 protein (F), adipocyte gene expression (G), Oil red O staining (H), Alizarin red-S staining (I), and osteoblast gene expression (J) in MSCs expressing NT Ctrl or VCAM1 shRNAs cocultured with ARP-1 or MM.1S cells in 1:1 OB:AD medium. Blot in (F) is a representative of three independent experiments, and blot quantitation is presented in fig. S3B. Data are means SD from n = 3 independent experiments. *P 0.05 and **P 0.01. P values were determined using one-way ANOVA with Tukeys multiple comparisons test except in (G) and (J), where Students t test for paired samples were used.

Because VCAM1 stimulates intracellular signaling that results in the activation of protein kinase C (PKC), we examined PKC activation in cocultures. Coculture of myeloma cells and MSCs enhanced the phosphorylation of PKC1 but did not affect phosphorylation of the PKC isoforms PKC, PKC, or PKC/ or the abundance of total PKC and reduced the phosphorylation of PKC and PKC (Fig. 6, A and B). Addition of the PKC inhibitor Go6976 to the cocultures markedly reduced PKC1 phosphorylation and PPAR2 protein in MSC cells cocultured with ARP-1 or MM.1S cells (Fig. 6C and fig. S4). Functionally, treatment of cocultures with Go6976 reduced Oil red O staining and increased Alizarin red-S staining (Fig. 6, D to F). Together, these results demonstrate that myeloma cells activated PPAR2 in MSCs and induced MSC differentiation into adipocytes rather than osteoblasts through the integrin 4-VCAM1-PKC1 pathway.

(A) Western blotting for all phosphorylated PKCs (p-PKC pan), the indicated phosphorylated PKC isoforms, and total PKC in MSCs cultured alone or cocultured with ARP-1 or MM.1S myeloma cells. The abundances of total PKC served as protein loading controls. (B) Quantification of the phosphorylation of PKC isoforms in MSCs cocultured with myeloma cells in (A) relative to the MSC-only control. The cutoff values are fold change more than twofold or less than 0.5-fold. (C) Western blotting for phosphorylated PKC1, total PKC, and PPAR2 in MSCs cocultured with ARP-1 or MM.1S cells in the presence of the PKC inhibitor Go6976 or DMSO (control). Actin is a loading control. Blot is a representative of three independent experiments, and blot quantitation is presented in fig. S4. (D) Representative images of Oil red O staining and Alizarin red-S staining of MSCs cultured alone or cocultured with ARP-1 or MM.1S myeloma cells in the presence of the PKC inhibitor Go6976 or DMSO (control). Scale bar, 5 mm. (E and F) Quantification of Oil red O staining (E) and Alizarin red-S staining (F), in cells treated as in (D). Data are means SD from n = 3 independent experiments. *P 0.05 and **P 0.01. P values were determined using one-way ANOVA with Tukeys multiple comparisons test.

Because a key mechanism of regulation of PPAR2 is its ubiquitylation-dependent proteasome-mediated degradation (16), we added the proteasome inhibitor MG132 to cultures of MSCs. We found that treatment with MG132 increased the presence of PPAR2 protein in MSCs in a time- and dose-dependent manner (Fig. 7A and fig. S5A). MG132 treatment causes the accumulation of ubiquitylated PPAR2 in MSCs, and coculturing these cells with myeloma cells reduced PPAR2 ubiquitylation (Fig. 7B and fig. S5B). However, the addition of a neutralizing antibody against VCAM1 to the cocultures restored ubiquitylation of PPAR2 (Fig. 7C and fig. S5C). These results suggested that myeloma cells activate PPAR2 in MSCs through inhibition of its ubiquitylation.

(A) Western blotting analysis for PPAR2 in MSCs cultured in 1:1 OB:AD medium and treated with the proteasome inhibitor MG132 for the indicated amounts of time. Actin is a loading control. (B) Immunoblotting (IB) for ubiquitin in PPAR2 immunoprecipitates (IP) from MSCs cultured alone or cocultured with ARP-1 or MM.1S myeloma cells in the presence of MG132. (C) Western blotting for ubiquitin in PPAR2 immunoprecipitates from MSCs cocultured with ARP-1 or MM.1S cells in the presence of MG132 and an antibody against VCAM1 or IgG (control). (D) Expression of the E3 ligaseencoding genes USP7, MURF1, MKRN1, CRBN, CRL4B, and TRIM23 in MSCs cocultured with myeloma cells relative to the expression in MSCs cultured alone (No MM). Data are means SD from n = 3 independent experiments. **P 0.01. P values were determined using one-way ANOVA with Tukeys multiple comparisons test. (E) Western blotting for USP7, MURF1, and MKRN1 in MSCs cultured alone or cocultured with myeloma cells. (F) Western blotting for MURF1 in MSCs cocultured with ARP-1 or MM.1S myeloma cells and treated with Go6976 or DMSO (control) as indicated. (G) Immunoblotting for MURF1 or PPAR2 in PPAR2 or MURF1 immunoprecipitates, respectively, from MSCs. IgG immunoprecipitates and whole-cell lysate (input) were used as controls. (H) Immunoblotting for ubiquitin in PPAR2 immunoprecipitates from MSCs expressing nontarget control (NT Ctrl) or MURF1 shRNAs in the presence of MG132. Each blot is representative of n = 3 independent experiments, and blot quantitation is presented in fig. S5.

To investigate the mechanism by which myeloma cells inhibited PPAR2 ubiquitylation, we examined the E3 ubiquitin ligases known to induce ubiquitylation of PPARs (17). Among the tested ligases, we found that MURF1 mRNA (Fig. 7D) and MURF1 protein (Fig. 7E and fig. S5D) were reduced in MSCs cocultured with myeloma cells. Addition of the PKC inhibitor Go6976 to the cocultures increased MURF1 protein in MSCs (Fig. 7F and fig. S5E), indicating that myeloma cells inhibited MURF1 production in MSCs through the PKC signaling pathway. Because the effects of MURF1 on PPAR2 ubiquitylation are unclear, we examined the interaction of these two proteins in MSCs. Co-immunoprecipitation of PPAR2 from MSCs demonstrated an interaction between MURF1 and PPAR2 (Fig. 7G), and knockdown of MURF1 in MSCs reduced the ubiquitylation of PPAR2 (Fig. 7H and fig. S5, F and G). These results demonstrate that myeloma cells activated PPAR2 in MSCs by reducing MURF1-mediated ubiquitylation of PPAR2.

To test the influence of myeloma cells on MSC differentiation in vivo, we established an extramedullary bone formation model in mice. Matrigel containing MSCs and Matrigel containing MSCs plus -irradiated ARP-1 cells were subcutaneously implanted into the right and left flanks of nonobese diabetic/severe combined immunodeficiency/interleukin-2rnull mice, respectively (Fig. 8A). Each sample also included human endothelial colony-forming cells (ECFCs) to stimulate blood vessel formation in the implant. In line with results of a previous study (18), we observed lower bone density in the extramedullary bones that formed in the left flanks, which were implanted with MSCs plus irradiated myeloma cells, compared to the extramedullary bones that formed on the right side, which were implanted with MSCs alone (Fig. 8A). Furthermore, we examined subcutaneous tissues on both sides of mice using histologic or immunohistochemical staining with antibodies against the mature osteoblast marker osteocalcin, the adipocyte marker perilipin, the myeloma marker CD138, and human MURF1. We observed lower numbers of new bones and osteocalcin+ osteoblasts and higher numbers of perilipin+ adipocytes in tissues on the sides of mice implanted with both MSCs and myeloma cells, reduction of MURF1 abundance in tissues on the sides of mice implanted with MSCs alone, and CD138+ cells only in tissues on the sides of mice implanted with myeloma cells (Fig. 8B).

(A) Representative images of subcutaneous tissues and bone density in mice implanted with human MSCs plus ECFCs in the right flank and MSCs plus ECFCs mixed with ARP-1 myeloma cells in the left flank. The arrows indicate bone formation in subcutaneous tissue, and the bars indicate bone density. (B) Representative hematoxylin and eosin (H&E) and immunohistochemical staining for the osteoblast marker osteocalcin, the adipocyte marker perilipin, the myeloma cell marker CD138+, and MURF1 of the subcutaneous tissues from (A). Scale bar, 20 m. Data represent n = 3 independent experiments with five mice each. (C) Expression of MURF1 in MSCs from BM aspirates from 12 patients with myeloma and 12 age-matched healthy donors relative to expression in a randomly selected sample from healthy donor. Data are from n = 3 experiments using the same donor source material. *P 0.05. P values were determined using Students t test. (D and E) Western blotting for MURF1 and PPAR2 (D) and Alizarin red-S and Oil red O staining (E) in MSCs from BM aspirates from three healthy donors and three patients with myeloma. Blots and images are representative of three experiments using the same donor materials, and blot quantitation is presented in fig. S6. Scale bars, 5 mm (whole wells) and 100 m (enlargements). (F) Quantitation of Alizarin red-S and Oil red O staining in the cultures of MSCs from BM aspirates from healthy donors and patients with myeloma in (C). Data are from n = 3 experiments using the same donor source material. P values were determined using Students t test. OD490, optical density at 490 nm.

We also isolated MSCs from the BM of 12 healthy human donors and 12 age-matched patients with myeloma and found markedly lower MURF1 mRNA expression in patient-derived MSCs compared to healthy donor MSCs (Fig. 8C). Western blotting validated the negative correlation between MURF1 and PPAR2 at the protein level in MSCs isolated from 3 of 12 samples in both groups (Fig. 8D and fig. S6). When we cultured these primary MSCs in 1:1 AD:OB medium, we found lower Alizarin red-S staining and higher Oil red O staining in cultures of patient-derived MSCs than in cultures of healthy donor MSCs (Fig. 8, E and F). These findings demonstrate that myeloma cells reduced MURF1 in MSCs and skewed MSC differentiation to favor adipogenesis, resulting in the suppression of osteoblast-mediated new bone formation in myeloma-bearing mice and in cells from patients with myeloma.

Using scRNA-seq, an in vitro coculture system, and mouse models, we demonstrated that myeloma cells shift the differentiation of MSCs into adipocytes rather than osteoblasts. Mechanistic studies revealed that integrin 4 on myeloma cells bound to VCAM1 on MSCs and inhibited ubiquitylation of PPAR2 through PKC-MURF1 signaling. The resulting increase in PPAR2 enhanced adipogenesis and suppressed osteoblastogenesis from MSCs. Thus, our study elucidates a previously unknown mechanism underlying myeloma-induced suppression of osteoblast-mediated bone formation and provides a potential approach for treating bone resorption in patients with myeloma.

Suppressed differentiation of osteoblasts is well known to be a key reason for bone loss and skeleton-related events in patients with myeloma (19). The molecules and pathways involved in myeloma-induced suppression of osteoblastogenesis include the Wnt signaling inhibitor Dickkopf-related protein 1 (DKK-1) (2, 20). However, antibody-mediated blocking of DKK-1 function cannot restore new bone formation completely or heal myeloma-induced resorbed bone, suggesting that additional factors expressed by myeloma cells critically affect bone formation. In the present study, we demonstrated that the 4 subunit of integrin, which is highly abundant in myeloma cells, promoted MSC differentiation into adipocytes, demonstrating that adhesion moleculesbut not soluble factorsproduced by myeloma cells primarily mediated the shift from osteoblastogenesis to adipogenesis. Integrin 41, also known as very late antigen-4, is a cell surface heterodimer present on malignant cells in patients with many types of cancer, including myeloma (21). It is a key adhesion molecule that acts as a receptor for the extracellular matrix protein fibronectin and the cellular receptor VCAM1. Interaction between integrin 41 and VCAM1 can activate mature osteoclast formation in patients with bone-metastatic breast cancer (22). In patients with multiple myeloma, this interaction promotes the secretion of interleukin-7 by tumor cells, which inhibits the expression of RUNX-2, which encodes a transcription factor that is essential for osteoblast differentiation, and RUNX-2 transcriptional regulatory activity in MSCs (23). This interaction also increases DKK-1 secretion by myeloma cells. Adding to these known mechanisms, we revealed that binding of myeloma cell integrin 41 to VCAM1 on the MSC surface activated the PKC signaling pathway. We also identified activation of PKC1, suppression of the downstream mediator MURF1, and the fundamental roles of such signaling pathways in the promotion of the MSC-derived adipocyte lineage. PKCs are also reportedly associated with Jagged-Notch signaling pathways, and they can regulate the transition of embryonic stem cells differentiating into postmitotic neurons (24). Some immunomodulatory drugs, such as lenalidomide, may affect osteoblast differentiation through this pathway (25), indicating the important role of Jagged-Notch in osteoblast differentiation from MSCs. We may further investigate their impacts and mechanisms on myeloma-induced the shift of MSC fates in our next studies.

BM adipocytes are recognized as important regulators of bone remodeling rather than just being inert filler cells (26, 27). Normal BM adipocytes have been shown to be reprogrammed by myeloma cells and gain the ability to resorb bone in myeloma patients in remission (13). Focusing on the determination of MSC fate in this study, we investigated the molecular mechanism underlying the shift from osteoblastogenesis to adipogenesis induced by myeloma cells. Lineage-tracing experiments have revealed that adipocytes can also originate from osterix-positive cells and are closely related to osteoblasts (28). Chan et al. (29) reported that BM adipocytes were derived from a progenitor cell that was also the progenitor for osteoblasts. In addition, Gao et al. (30) reported plasticity between BM adipocytes and osteoblasts and potential transdifferentiation and transformation between these two identities after initiating differentiation. Despite this new knowledge about the balance between osteoblastogenesis and adipogenesis, how myeloma cells regulate this balance and transformation of MSCs is still unclear.

scRNA-seq can identify subpopulations using the transcriptome to avoid the complicated isolation procedures after cell-cell contact culture (15). We found that MSCs could be naturally divided into two populations by transcriptomic data, and at least one cluster of MSCs cocultured with myeloma cells highly expressed adipocyte marker genes. Coculture of myeloma cells pushed MSC differentiation toward adipocytes rather than osteoblasts, resulting in the suppression of bone formation in the in vivo extramedullary bone assay. Because MSCs are pluripotent stem cells capable of differentiation into other cell types, such as chondrocytes and skeletal muscle cells (31), whether myeloma cells affect MSC transformation into these cell types instead of osteoblasts remains unclear. It is possible that the observed differentiation from MSC to adipocyte in the presence of myeloma cells might have been rather the result of a differentiation of MSCs into osteoblasts followed by a transdifferentiation from osteoblast into adipocyte. Further investigation is needed to address this possibility.

Like other transcription factors and coregulators, PPAR2 can undergo posttranslational modifications, such as phosphorylation, acetylation, and SUMOylation (32). Researchers have identified the key enzymes and target amino acid sites involved in these modifications, but modification of PPAR2 by ubiquitylation, especially that induced by myeloma cells, is still unclear. Many E3 ligases, such as MURF1 and makorin ring finger protein 1 (MKRN1), are reported to be regulators of ubiquitylation of PPAR proteins (17, 3335), whereas investigators have identified only polyubiquitylation at Lys184 and Lys185 (K184/185) mediated by MKRN1 (16). In the present study, we demonstrated that the E3 ligase MURF1 contributed to PPAR2 ubiquitylation, and inhibition of MURF1 by myeloma cells reduced PPAR2 ubiquitylation, leading to enhanced protein stability in MSCs. MURF1 contains a canonical N-terminal RING-containing E3 ligase that is required for its ubiquitin ligase activity (36). Others have reported dysregulation of MURF1 in experimental models of fasting, diabetes, cancer, denervation, and immobilization (37). However, none have reported the substrate proteins, such as PPAR2, that are targeted for proteasomal degradation by MURF1 in patients with myeloma bone disease. Although the amino acids in PPAR2 that MURF1 targets remain to be identified, we demonstrated that the reduced MURF1 production in MSCs induced by myeloma cells was critical for the inhibition of PPAR2 ubiquitylation and thus stabilization of the PPAR2 protein. Other posttranslational modifications may also regulate PPAR2 protein, especially SUMOylation, which was not addressed in the current study. For example, the transcriptional activity of PPAR2 can be inhibited by SUMOylation at Lys107 to regulate insulin sensitivity (38), and growth differentiation factor 11 promotes osteoblastogenesis through enhancement of PPAR2 SUMOylation (39). A logical next step could be the investigation of the role of SUMOylation in myeloma-induced MSC transformation and how it interplays with the mechanisms described here.

In summary, our results shed light on the cross-talk between myeloma cells and MSCs and the impact of this interaction on the determination of the MSC-derived adipocyte lineage and the suppression of osteoblastogenesis from MSCs. Myeloma cell integrin 4 promoted phosphorylation of PKC1 through VCAM1, and the activated PKC1 reduced the production of MURF1 in MSCs, leading to reduced PPAR2 ubiquitylation. Therefore, counteracting 4-VCAM1-MURF1mediated adipogenesis from MSCs may be a promising strategy to heal myeloma-induced bone resorption.

Myeloma cell lines ARP-1 and ARK were provided by University of Arkansas for Medical Sciences (Little Rock, AR, USA), and others were purchased from American Type Culture Collection. Primary myeloma cells or normal plasma cells were isolated from the BM aspirates of patients with myeloma or healthy donors using antibody-coated magnetic beads against CD138, respectively (Miltenyi Biotec Inc.) (40). The cells were maintained in RPMI 1640 medium with 10% fetal bovine serum (FBS). MSCs from BM of healthy donors or patients with myeloma were maintained and augmented in Dulbeccos modified Eagles medium (DMEM) with 10% FBS (13). Information of healthy donors and patients were listed in table S1. The study was approved by the Institutional Review Board at The University of Texas MD Anderson Cancer Center.

Human MSCs were generated from BM mononuclear cells from fetal bones of healthy human donors, characterized using flow cytometry, and labeled with antibodies against MSC markers (CD44, CD90, and CD166) (41). Mature adipocytes were generated from MSCs using an adipocyte medium, which was formulated of DMEM medium with 10% FBS, 1 M dexamethasone, 0.2 mM indomethacin, insulin (10 g/ml), and 0.5 mM 3-isobutyl-l-methylxanthine (41). Mature adipocytes were fixed with 4% paraformaldehyde, stained with Oil red O for 1 hour, and observed under a light microscope. Mature osteoblasts were generated from MSCs using an osteoblast medium, which was formulated of alpha MEM medium with 10% FBS, 100 nM dexamethasone, 10 mM -glycerophosphate, and 0.05 mM l-ascorbic acid 2-phosphate (42). The bone-forming activity of osteoblasts was determined using Alizarin red-S staining (43, 44). Human MSCs were cultured alone or cocultured with myeloma cells at a ratio of 5:1 in MSC medium, osteoblast medium, adipocyte medium, or 1:1 mixed of osteoblast and adipocyte medium with or without inhibitors (G3335 or Go6976) or neutralizing antibodies for 2 weeks. Addition of dimethyl sulfoxide (DMSO) served as vehicle control for inhibitor-treatment experiments, and addition of IgG served as control for antibody-neutralizing experiments. In the transwell nondirect contact model, adipocytes were seeded onto the bottom of culture wells and cocultured with the myeloma cells on the insert. In direct contact coculture system, MSCs were seeded together with the myeloma cells in the culture wells to allow direct cell-cell contact. Supernatants collected from 24-hour cultures of myeloma cells were added to the MSCs in mixed osteoblast and adipocyte medium at a ratio of 1:5. In the experiments with primary cells, MSCs were cultured in the mixed medium for a week (45) and then cocultured with primary myeloma cells isolated from BM aspirates from patients with myeloma or normal plasma cells from BM of healthy donors for another week. Medium, inhibitors, and antibodies were refreshed every 3 days. After culture, the myeloma cells were removed, and the residual cells were stained with Alizarin red-S to assess osteoblast differentiation and with Oil red O to assess adipocyte differentiation. Culture of MSCs alone served as a control.

Single-cell preparation, complementary DNA (cDNA) library synthesis, RNA sequencing, and data analysis were performed by Gene Denovo Inc. Briefly, 1 106 MSCs were plated for 6 hours, 5 106 myeloma cells were added to the MSCs directly, and the cells were cocultured in mixed culture media for 48 hours; control MSC cells were cultured alone at the same media and then mixed with myeloma cells at the same ratio just before preparation for analysis. After removal of dead cells, the cells in these groups were counted using a Countess II Automated Cell Counter, and the concentration was adjusted to 1000 cells/l. The single-cell suspensions were bar-coded labeled and reverse-transcribed into scRNA-seq library using the Chromium Single Cell 3 GEM, Library and Gel Bead Kit (10X Genomics). The cDNA libraries from two independent experiments were sequenced on the Illumina HiSeq X-Ten platform, and data were pooled for the analysis. Myeloma cells were excluded using CD138 markers. The raw scRNA-seq data were aligned, filtered, and normalized using Cell Ranger (10X Genomics) software (tables S2 to S6). The cell subpopulation was grouped by graph-based clustering based on the gene expression profile of each cells in Seurat (tables S7 and S8). Subsequent data analysis including standardization, cell subpopulation, difference of gene expression, and marker gene screening were achieved by Seurat software.

MSCs were cultured alone or cocultured with myeloma cells with or without G3335 or neutralizing antibodies for 48 hours. In some experiments, MG132 was added to the cultures 6 hours before the cell collection. Addition of DMSO served as vehicle control for inhibitor experiment; addition of IgG served as neutralizing antibody control.

Quantitative real-time PCR was performed as described (46). The primers are listed in table S9. For Western blotting, cells were lysed with 1 lysis buffer (Cell Signaling Technology), subjected to 4 to 20% gradient gel electrophoresis, transferred to, and immunoblotted with antibodies against integrin 4 (R&D Systems), integrin 1, VCAM1, PKC, MURF1, and phosphorylated isoforms of PKC along with p-PKC-pan (Cell Signaling Technology) and PPAR2 (Santa Cruz Biotechnology). The membrane was stripped and reprobed with an antibody against -actin to ensure equal protein loading, and last, signals were detected using peroxidase-conjugated secondary antibody followed by enhanced chemiluminescence system (Millipore) in the MiniChem system (Saizhi Biotech), and quantitative analysis of blots were performed using the Fiji-based ImageJ software (version 1.51n, National Institutes of Health, Bethesda, MA, USA).

Viral particles were produced by human embryonic kidney 293T cells transfected with PMD2G and PSPAX2 packaging plasmids (Addgene) together with lentivirus-expressing shRNA vectors targeting 4, MURF1, or VCAM1 (Sigma-Aldrich). Nontargeted shRNA control (Sigma-Aldrich) was used as control. Sequences for knocking down specific genes are the following: 4, 5-CCGGGCTCCGTGTTATCAAGATTATCTCGAGATAATCTTGATAACACGGAGCTTTTT-3; VCAM1, 5-CCGGGGAATTAATTATCCAAGTTACCTCGAGGTAACTTGGATAATTAATTCCTTTTTTG-3; MURF1, 5-CCGGGAAGAGGAAGAGTCCACAGAACTCGAGTTCTGTGGACTCTTCCTCTTCTTTTTG-3 or 5-CCGGGTATAATAATGCCTGGTCATTCTCGAGAATGACCAGGCATTATTATACTTTTTG-3. Supernatants carrying the viral particles were harvested 48 hours later and concentrated to a 100 volume using polyethylene glycol 8000 (Sigma-Aldrich). MSCs (1 106 cells) were seeded 6 hours before the infection. Concentrated viral particles were added to MSCs or myeloma cells, respectively, in the presence of polybrene (8 g/ml) for 12 hours. The medium was then changed, and cells were cultured for another 48 hours until further management.

Cells were harvested and lysed using NP-40 lysis buffer supplemented with complete protease inhibitors, and the supernatant was precleaned with protein G beads (Thermo Fisher Scientific) and incubated with a mouse antibody against MURF1 (Santa Cruz Biotechnology) or monoclonal rabbit antibody against PPAR2 antibody (Santa Cruz Biotechnology) at 4C overnight with protein A/G agarose beads (Thermo Fisher Scientific). The next day, the pellet was washed four times with lysis buffer and then subjected to Western blot analysis using the antibodies against PPAR2 or MURF1. IgG was used as a control and total cell lysates (5%) were used as input controls.

For a ubiquitylation assay, diluted lysates were incubated with an antibody against PPAR2 at 4C overnight after precleaning with protein G beads (Thermo Fisher Scientific). Protein G beads were added to the washed lysate/antibody mixture at 4C for 4 hours. The resin was washed and applied to Western blot analysis using an antibody against ubiquitin.

MSCs were cultured alone or cocultured with myeloma cells for 2 weeks. Abundance of FABP4 and osteocalcin was assessed by immunofluorescence using fluorescein isothiocyanate or allophycocyanin-conjugated antibodies (BD Biosciences). After staining, cells were resuspended in phosphate-buffered saline with 1% FBS and analyzed using a BD LSR Fortessa flow cytometer.

The animal experiments in the present study were approved by the MD Anderson Institutional Animal Care and Use Committee. In vivo extramedullary bone formation in nonobese diabetic/severe combined immunodeficiency/interleukin-2rnull mice was established and examined (18). Briefly, MSCs alone or a mixture of human MSCs (1.5 106) and human ECFCs (1.5 106) in 0.2 ml of Matrigel (Corning Inc.) was subcutaneously injected into the right flanks of mice. This mixture and an additional 2 105 -irradiated (5000 centigrays) myeloma cells were injected into the left flanks of the mice. At 8 weeks after implantation, subcutaneous tissues were established, and the mice were intraperitoneally injected with OsteoSense 750 to assess new bone formation in those tissues. The subcutaneous tissues were collected after the mice were sacrificed and subjected to hematoxylin and eosin or immunohistochemical staining of cells labeled with an antibody against osteocalcin (a marker of mature osteoblasts), an antibody against perilipin (a marker of mature adipocytes), or an antibody against CD138 (a marker of myeloma cells).

The subcutaneous tissues were extracted from the mice and then formalin-fixed and paraffin-embedded. Tissue sections were deparaffinized with xylene and rehydrated to water through a graded alcohol series. Endogenous peroxidase activity was quenched with 3% hydrogen peroxide. The presence of CD138 (R&D Systems), osteocalcin, perilipin, and MURF1 (Abcam) in tissues was detected using specific antibodies. Signals were detected using secondary biotinylated antibodies and streptavidin/horseradish peroxidase. Chromagen 3,3-diaminobenzidine/H2O2 (Dako) was used, and slides were counterstained with hematoxylin. All slides were observed under a light microscope, and images were captured using a SPOT RT camera (Diagnostic Instruments).

Experimental values were expressed as means SD unless indicated otherwise. Statistical significance was analyzed using the GraphPad Prism v7.0 with two-tailed unpaired Students t tests for comparison of two groups and one-way analysis of variance (ANOVA) with Tukeys multiple comparisons test for comparison of more than two groups. P values less than 0.05 were considered statistically significant. All results were reproduced in at least three independent experiments.

stke.sciencemag.org/cgi/content/full/13/633/eaay8203/DC1

Fig. S1. G3335 inhibits PPAR2 accumulation in MSCs cocultured with myeloma cells.

Fig. S2. 4 KD in myeloma cells.

Fig. S3. VCAM1 knockdown in MSCs.

Fig. S4. PKC inhibition reduces PKC1 phosphorylation and PPAR2 abundance in MSCs cocultured with myeloma cells.

Fig. S5. Coculture with myeloma cells reduces ubiquitylation of PPAR2 in MSCs.

Fig. S6. MSCs from patients with myeloma show decreased MURF1 and increased PPAR2.

Table S1. Characteristics of patients with myeloma and healthy donors.

Table S2. Read quality control of the samples for scRNA-seq.

Table S3. Mapping quality control of aligned scRNA-seq data.

Table S4. Basic information of the aggregated samples for scRNA-seq before and after normalization.

Table S5. Information of each sample after aggregation.

Table S6. Cell quality control showing the cell numbers before and after the filtration.

Table S7. Number of cells in each subpopulation.

Table S8. Number of cells in each subpopulation of control and cocultured samples.

Table S9. Primers used in the quantitative reverse transcription PCR analysis.

Data file S1. scRNA-seq data from control sample.

Data file S2. scRNA-seq data from coculture sample.

Acknowledgments: We thank M. J. Li from Department of Genetics, Tianjin Medical University for the evaluation of our statistical analysis. Funding: This work was supported by R01 grants from NCI (CA190863 and CA193362 to J.Y.) and by the Research Scholar Grant from the American Cancer Society (127337-RSG-15-069-01-TBG to J.Y.). It was also supported by NIH/NCI (Core Labs at UT MD Anderson Cancer Center, P30CA016672) for the Small Animal Imaging and Research Histopathology Facilities. Author contributions: Z.L. and J.Y. designed all experiments and wrote the manuscript. H.L., Z.L., and J.H performed all experiments and statistical analysis. P.L. provided and interpreted patient samples. Q.T. provided critical suggestions. Conflict of interests: The authors declare that they have no competing interests. Data and materials availability: All of the data needed to evaluate the conclusions in the paper are provided in the main text or the Supplementary Materials. Stable cell lines carrying targeted shRNA are available with a materials transfer agreement between Houston Methodist Research Institute and the requesting institution.

View original post here:
Myeloma cells shift osteoblastogenesis to adipogenesis by inhibiting the ubiquitin ligase MURF1 in mesenchymal stem cells - Science

ViaCyte to Present at the Jefferies Virtual Healthcare Conference – Monterey County Weekly

SAN DIEGO, May 28, 2020 /PRNewswire/ -- ViaCyte, Inc., a privately held regenerative medicine company, announced today that ViaCyte's President and CEO Paul Laikind, Ph.D. is scheduled to present at the Jefferies Virtual Healthcare Conference on Thursday, June 4, 2020 at 10 a.m. Eastern Time in Track 9. The company recently announced a $27 million financingto advance next generation cell therapies for diabetes. ViaCyte is the first company to demonstrate production of C-peptide, a biomarker for insulin, in patients with type 1 diabetes receiving a stem cell-derived islet replacement.

During the presentation, Dr. Laikind will discuss ViaCyte's leadership in discovering and developing novel cell replacement therapies to treat human diseases. The company has two product candidates, PEC-Direct and PEC-Encap (also known as VC-01), in clinical trials. Both have the potential to transform the way type 1 diabetes is managed to the extent of providing a functional cure for the disease. With CRISPR Therapeutics, ViaCyte is developing immune-evasive stem cell lines from its proprietary CyT49 pluripotent stem cell line. These immune-evasive stem cell lines, which are being used in the PEC-QT program, have the potential to further broaden the availability of cell therapy for all patients with insulin-requiring diabetes, type 1 and type 2, as well as other potential indications. The Company's leadership is reflected in its robust intellectual property portfolio, which includes hundreds of issued patents and pending applications worldwide.

The live audio webcast of the company's presentation will be available to conference registrants.

About ViaCyte

ViaCyte is a privately held regenerative medicine company developing novel cell replacement therapies as potential long-term diabetes treatments to achieve glucose control targets and reduce the risk of hypoglycemia and diabetes-related complications. ViaCyte's product candidates are based on directed differentiation of pluripotent stem cells into PEC-01 pancreatic islet progenitor cells, which are then implanted in durable and retrievable cell delivery devices. Over a decade ago, ViaCyte scientists were the first to report on the production of pancreatic cells from a stem cell starting point and the first to demonstrate in an animal model of diabetes that, once implanted and matured, these cells secrete insulin and other pancreatic hormones in response to blood glucose levels and can be curative. More recently, ViaCyte demonstrated that when effectively engrafted, PEC-01 cells can mature into glucose-responsive insulin-producing cells in patients with type 1 diabetes. To accelerate and expand its efforts, ViaCyte has established collaborative partnerships with leading companies including CRISPR Therapeutics and W.L. Gore & Associates. ViaCyte is funded in part by the California Institute for Regenerative Medicine (CIRM) and JDRF. ViaCyte is headquartered in San Diego, California. For more, please visit http://www.viacyte.comand connect with ViaCyte on Twitter, Facebook, and LinkedIn.

View post:
ViaCyte to Present at the Jefferies Virtual Healthcare Conference - Monterey County Weekly

COVID-19 Impact on Global Cancer Stem Cell Therapy Market 2020: Industry Trends, Size, Share, Growth Applications, SWOT Analysis by Top Key Players…

The market is primarily driven by increasing number of cancer patients suffering from many different types of cancer. In addition, stem cell therapy is a painless and incision less treatment procedure is likely to boost the market growth. However, lack of suitable and donor matching sample might restrict the market growth.

Report:www.orianresearch.com/requestle/1040620

Development policies and plans are discussed as well as manufacturing processes and cost structures are also analyzed. This report also states import/export consumption, supply and demand Figures, cost, price, revenue and gross margins.

Top Key Companies Analyzed inGlobal Cancer Stem Cell Therapy Market are Thermo Fisher Scientific, Merck Kgaa, Bionomics, Lonza, Stemline Therapeutics, Miltenyi Biotec, Promocell, Macrogenics, Oncomed Pharmaceuticals and Irvine Scientific

Key Benefit of This Report:

Global Cancer Stem Cell Therapy Industry 2019 Market Research Report is spread across 121 pages and provides exclusive vital statistics, data, information, trends and competitive landscape details in this niche sector.

Report:www.orianresearch.com/040620

Target Audience:

Research Methodology:

The market is derived through extensive use of secondary, primary, in-house research followed by expert validation and third party perspective, such as, analyst reports of investment banks. The secondary research is the primary base of our study wherein we conducted extensive data mining, referring to verified data sources, such as, white papers, government and regulatory published articles, technical journals, trade magazines, and paid data sources.

For forecasting, regional demand & supply factors, recent investments, market dynamics including technical growth scenario, consumer behavior, and end use trends and dynamics, and production capacity were taken into consideration.

Different weightages have been assigned to these parameters and quantified their market impacts using the weighted average analysis to derive the market growth rate.

The market estimates and forecasts have been verified through exhaustive primary research with the Key Industry Participants (KIPs), which typically include:

Report:www.orianresearch.com/checkout/1040620

Major Points Covered in Table of Contents:

1 Introduction

2 Research Methodology

3 Executive Summary

4 Global Cancer Stem Cell Therapy Market Overview

5 Global Cancer Stem Cell Therapy Market, by Product Type

6 Global Cancer Stem Cell Therapy Market, by Application

7 Global Cancer Stem Cell Therapy Market by Region

8 Competitive Landscape

9 Company Profiles

10 Key Insights

Customization Service of the Report:Orian Research provides customisation of reports as per your need. This report can be personalised to meet your requirements. Get in touch with our sales team, who will guarantee you to get a report that suits your necessities.

About Us:Orian Research is one of the most comprehensive collections of market intelligence reports on the World Wide Web. Our reports repository boasts of over 500000+ industry and country research reports from over 100 top publishers. We continuously update our repository so as to provide our clients easy access to the worlds most complete and current database of expert insights on global industries, companies, and products. We also specialize in custom research in situations where our syndicate research offerings do not meet the specific requirements of our esteemed clients.

Contact Us:Ruwin MendezVice President Global Sales & Partner RelationsOrian Research ConsultantsUS: +1 (832) 380-8827 | UK: +44 0161-818-8027Email: [emailprotected]Website: http://www.orianresearch.com/

Continued here:
COVID-19 Impact on Global Cancer Stem Cell Therapy Market 2020: Industry Trends, Size, Share, Growth Applications, SWOT Analysis by Top Key Players...

AdventHealth launches first-of-its-kind immunotherapy treatment, clinical trial for specific blood cancer patients – The Daily Ridge

AdventHealth launches first-of-its-kind immunotherapy treatment, clinical trial for specific blood cancer patients

The innovative treatment being tested, which uses donated immune cells, offers a potential last chance of survival after conventional treatments have failed.

ORLANDO, Fla.,May 27, 2020 AdventHealth physicians are the first in the world to launch a pioneering treatment targeting certain blood cancers for patients who have exhausted all other types of therapy.Antigen-Specific T-Cell therapy, which is the subject of a clinical trial at AdventHealth Orlando, uses the immune cells to target cancer cells, and provides what is often the final treatment opportunity for people suffering from certain types of acute myeloid leukemia (AML) and myelodysplastic syndromes(MDS).The first patient to receive this therapy was treated in late April at AdventHealth Orlando and is recovering at home. He will be monitored to determine the efficacy of the treatment, which may take several months.More than 19,000 people will be diagnosed with AML this year in the United States, and over 11,000 people will die from the disease, according to the National Cancer Institute. The five-year survival rate for AML is 28.7 percent, while in comparison, the five-year survival rate for leukemia is 63.7 percent.The number of people diagnosed with MDS in the country each year is uncertain, but is estimated at 10,000 or higher, the American Cancer Society reports.The best attribute of an immunotherapy treatment like this one is that its a precise, customizable and personalized way to treat cancer for those who have no options left, said Dr. Juan Carlos Varela, hematology oncologist at AdventHealth and principal investigator of the trial. The relapse after traditional forms of treatment for these patients is around 40 percent. That relapse is the No. 1 cause of death for this patient population. Their options are very limited and theres an urgent need for potentially lifesaving treatment options like this one.Antigen-Specific T-Cells are made by removing white blood cells from a donor (who had previously donated stem cells to the patient), generating immune cells that are tumor-specific, and then infusing the generated cells back into the patients bloodstream. Antigen-Specific T-Cells are able to attack specific cancer cells.Being the first in the world to launch this therapy, and to have the lead investigator on our team, shows our commitment to personalized medicine, which is the future of cancer care, said Dr. Mark A. Socinski, executive medical director of the AdventHealth Cancer Institute. Were excited to bring this innovative therapy to our patients and allow them to access this potentially lifesaving treatment close to home.The Antigen-Specific T-Cell Therapy clinical trial and the Blood and Marrow Transplant program are made possible by the generous support of community donors, including the AdventHealth Foundation of Central Florida.

See the article here:
AdventHealth launches first-of-its-kind immunotherapy treatment, clinical trial for specific blood cancer patients - The Daily Ridge

Gamida looks to build out cell therapy infrastructure – Bioprocess Insider – BioProcess Insider

Gamida Cell will initiate a BLA submission for lead cell therapy omidubicel later this year and grow inhouse manufacturing capabilities to support production of cancer candidate GDA-201.

Israeli cell therapy company Gamida Cell raised $60 million (55 million) last week through a public share offering. The money will be used to support the approval application and manufacturing capabilities of its cell therapy products, the firm said on an investor call last week.

Lead candidate omidubicel formerly known as NiCord is a hematopoietic stem cell transplantation (HSCT), or bone marrow transplant, which recently met its primary endpoint in a Phase III study.

Image: iStock/Pablo_K

We have focused on working towards initiating a BLA submission on a rolling basis in the fourth quarter of this year, which will position us for a potential launch in the second half of 2021, Gamida CEO Julian Adams said on the call. We are also advancing key activities to bring omidubicel to patients following potential FDA approval.

The firm has been using Lonza to produce clinical material of omidubicel, with the contract development and manufacturing organization (CDMO) constructing dedicated suites at its site in Geleen, The Netherlands to support the candidates progression towards commercialization last year, but Adams said Gamida is also expanding its own production capabilities.

Work is ongoing to build out our manufacturing infrastructure both as Lonza and at our own facility to help ensure sufficient and reliable commercial supply. We are also working to develop comprehensive hospital services and patient assistance programs designed to seamlessly bring omidubicel to patients.

Gamida is also actively recruiting for medical affairs talent and reps to support a launch, now the financing has closed. It was dependent on the successful financing, so that we would have the wherewithal to continue to build out all of the infrastructure both for medical affairs, commercial and manufacturing.

Gamidas second candidate is GDA-201, an investigational, natural killer (NK) cell-based cancer immunotherapy in Phase I development in patients with non-Hodgkin lymphoma (NHL) and multiple myeloma.

Its based on the platform that brought us the omidubicel program and a lot of experience knowledge and relationships that weve made are ones that were leveraging for development of GDA-201, Gamidas chief medical officer Ronit Simantov told stakeholders. We continue to develop that program with development of cryopreserved products in our laboratories, and we will intend to bring that to a clinical study of company sponsored multi-center studies for patients next year.

But while Gamida will rely somewhat on Lonza for omidubicel, for GDA-201 the firm wants to manufacture fully inhouse.

We are undertaking to manufacture the NK GDA-201 product in our own facilities, said Adams, and the key advantage now that weve learned how to cryopreserve and recover NK activity is to turn that into a GMP process. So, theres still some process development going on, but were quite confident that we will achieve [this].

Go here to read the rest:
Gamida looks to build out cell therapy infrastructure - Bioprocess Insider - BioProcess Insider

Rheumatoid Arthritis Stem Cell Therapy Market to Register Substantial Expansion by Fact.MR – The Cloud Tribune

The global Rheumatoid Arthritis Stem Cell Therapy market study presents an all in all compilation of the historical, current and future outlook of the market as well as the factors responsible for such a growth. With SWOT analysis, the business study highlights the strengths, weaknesses, opportunities and threats of each Rheumatoid Arthritis Stem Cell Therapy market player in a comprehensive way. Further, the Rheumatoid Arthritis Stem Cell Therapy market report emphasizes the adoption pattern of the Rheumatoid Arthritis Stem Cell Therapy across various industries.Request Sample Reporthttps://www.factmr.com/connectus/sample?flag=S&rep_id=1001The Rheumatoid Arthritis Stem Cell Therapy market report highlights the following players:The global market for rheumatoid arthritis stem cell therapy is highly fragmented. Examples of some of the key players operating in the global rheumatoid arthritis stem cell therapy market include Mesoblast Ltd., Roslin Cells, Regeneus Ltd, ReNeuron Group plc, International Stem Cell Corporation, TiGenix and others.

The Rheumatoid Arthritis Stem Cell Therapy market report examines the operating pattern of each player new product launches, partnerships, and acquisitions has been examined in detail.Important regions covered in the Rheumatoid Arthritis Stem Cell Therapy market report include:

North America (U.S., Canada)Latin America (Mexico, Brazil)Western Europe (Germany, Italy, U.K., Spain, France, Nordic countries, BENELUX)Eastern Europe (Russia, Poland, Rest Of Eastern Europe)Asia Pacific Excluding Japan (China, India, Australia & New Zealand)JapanMiddle East and Africa (GCC, S. Africa, Rest Of MEA)

The Rheumatoid Arthritis Stem Cell Therapy market report takes into consideration the following segments by treatment type:

Allogeneic Mesenchymal stem cellsBone marrow TransplantAdipose Tissue Stem Cells

The Rheumatoid Arthritis Stem Cell Therapy market report contain the following distribution channel:

HospitalsAmbulatory Surgical CentersSpecialty ClinicsHave Any Query? Ask our Industry Experts-https://www.factmr.com/connectus/sample?flag=AE&rep_id=1001

Buy the report at a discounted rate!!! Exclusive offer!!!

The Rheumatoid Arthritis Stem Cell Therapy market report offers a plethora of insights which include:

Changing consumption pattern among individuals globally.Historical and future progress of the global Rheumatoid Arthritis Stem Cell Therapy market.Region-wise and country-wise segmentation of the Rheumatoid Arthritis Stem Cell Therapy market to understand the revenue, and growth lookout in these areas.Accurate Year-on-Year growth of the global Rheumatoid Arthritis Stem Cell Therapy market.Important trends, including proprietary technologies, ecological conservation, and globalization affecting the global Rheumatoid Arthritis Stem Cell Therapy market.

The Rheumatoid Arthritis Stem Cell Therapy market report answers important questions which include:

Which regulatory authorities have granted approval to the application of Rheumatoid Arthritis Stem Cell Therapy in Health industry?How will the global Rheumatoid Arthritis Stem Cell Therapy market grow over the forecast period?Which end use industry is set to become the leading consumer of Rheumatoid Arthritis Stem Cell Therapy by 2028?What manufacturing techniques are involved in the production of the Rheumatoid Arthritis Stem Cell Therapy?Which regions are the Rheumatoid Arthritis Stem Cell Therapy market players targeting to channelize their production portfolio?Get Full Access of the Report @https://www.factmr.com/report/1001/rheumatoid-arthritis-stem-cell-therapy-market

Pertinent aspects this study on the Rheumatoid Arthritis Stem Cell Therapy market tries to answer exhaustively are:

What is the forecast size (revenue/volumes) of the most lucrative regional market? What is the share of the dominant product/technology segment in the Rheumatoid Arthritis Stem Cell Therapy market? What regions are likely to witness sizable investments in research and development funding? What are Covid 19 implication on Rheumatoid Arthritis Stem Cell Therapy market and learn how businesses can respond, manage and mitigate the risks? Which countries will be the next destination for industry leaders in order to tap new revenue streams? Which new regulations might cause disruption in industry sentiments in near future? Which is the share of the dominant end user? Which region is expected to rise at the most dominant growth rate? Which technologies will have massive impact of new avenues in the Rheumatoid Arthritis Stem Cell Therapy market? Which key end-use industry trends are expected to shape the growth prospects of the Rheumatoid Arthritis Stem Cell Therapy market? What factors will promote new entrants in the Rheumatoid Arthritis Stem Cell Therapy market? What is the degree of fragmentation in the Rheumatoid Arthritis Stem Cell Therapy market, and will it increase in coming years?Why Choose Fact.MR?

Fact.MR follows a multi- disciplinary approach to extract information about various industries. Our analysts perform thorough primary and secondary research to gather data associated with the market. With modern industrial and digitalization tools, we provide avant-garde business ideas to our clients. We address clients living in across parts of the world with our 24/7 service availability.

Read the original here:
Rheumatoid Arthritis Stem Cell Therapy Market to Register Substantial Expansion by Fact.MR - The Cloud Tribune

On the Origins of Modern Biology and the Fantastic: Part 19 Nalo Hopkinson and Stem Cell Research – tor.com

She just wanted to be somewhere safe, somewhere familiar, where people looked and spoke like her and she could stand to eat the food. Midnight Robber by Nalo Hopkinson

Midnight Robber (2000) is about a woman, divided. Raised on the high-tech utopian planet of Touissant, Tan-Tan grows up on a planet populated by the descendants of a Caribbean diaspora, where all labor is performed by an all-seeing AI. But when she is exiled to Touissants parallel universe twin planet, the no-tech New Half-Way Tree, with her sexually abusive father, she becomes divided between good and evil Tan-Tans. To make herself and New Half-Way Tree whole, she adopts the persona of the legendary Robber Queen and becomes a legend herself. It is a wondrous blend of science fictional tropes and Caribbean mythology written in a Caribbean vernacular which vividly recalls the history of slavery and imperialism that shaped Touissant and its people, published at a time when diverse voices and perspectives within science fiction were blossoming.

Science fiction has long been dominated by white, Western perspectives. Vernes tech-forward adventures and Wells sociological allegories established two distinctive styles, but still centered on white imperialism and class struggle. Subsequent futures depicted in Verne-like pulp and Golden Age stories, where lone white heroes conquered evil powers or alien planets, mirrored colonialist history and the subjugation of non-white races. The civil rights era saw the incorporation of more Wellsian sociological concerns, and an increase in the number of non-white faces in the future, but they were often tokensparts of a dominant white monoculture. Important figures that presaged modern diversity included Star Treks Lieutenant Uhura, played by Nichelle Nichols. Nichols was the first black woman to play a non-servant character on TV; though her glorified secretary role frustrated Nichols, her presence was a political act, showing there was space for black people in the future.

Another key figure was the musician and poet Sun Ra, who laid the aesthetic foundation for what would become known as the Afrofuturist movement (the term coined by Mark Dery in a 1994 essay), which showed pride in black history and imagined the future through a black cultural lens. Within science fiction, the foundational work of Samuel Delany and Octavia Butler painted realistic futures in which the histories and cultural differences of people of color had a place. Finally, an important modern figure in the decentralization of the dominant Western perspective is Nalo Hopkinson.

A similarly long-standing paradigm lies at the heart of biology, extending back to Darwins theoretical and Mendels practical frameworks for the evolution of genetic traits via natural selection. Our natures werent determined by experience, as Lamarck posited, but by genes. Therefore, genes determine our reproductive fitness, and if we can understand genes, we might take our futures into our own hands to better treat disease and ease human suffering. This theory was tragically over-applied, even by Darwin, who in Descent of Man (1871) conflated culture with biology, assuming the Wests conquest of indigenous cultures meant white people were genetically superior. After the Nazis committed genocide in the name of an all-white future, ideas and practices based in eugenics declined, as biological understanding of genes matured. The Central Dogma of the 60s maintained the idea of a mechanistic meaning of life, as advances in genetic engineering and the age of genomics enabled our greatest understanding yet of how genes and disease work. The last major barrier between us and our transhumanist future therefore involved understanding how genes determine cellular identity, and as well see, key figures in answering that question are stem cells.

***

Hopkinson was born December 20, 1960 in Kingston, Jamaica. Her mother was a library technician and her father wrote, taught, and acted. Growing up, Hopkinson was immersed in the Caribbean literary scene, fed on a steady diet of theater, dance, readings, and visual arts exhibitions. She loved to readfrom folklore, to classical literature, to Kurt Vonnegutand loved science fiction, from Spock and Uhura on Star Trek, to Le Guin, James Tiptree Jr., and Delany. Despite being surrounded by a vibrant writing community, it didnt occur to her to become a writer herself. What they were writing was poetry and mimetic fiction, Hopkinson said, whereas I was reading science fiction and fantasy. It wasnt until I was 16 and stumbled upon an anthology of stories written at the Clarion Science Fiction Workshop that I realized there were places where you could be taught how to write fiction. Growing up, her family moved often, from Jamaica to Guyana to Trinidad and back, but in 1977, they moved to Toronto to get treatment for her fathers chronic kidney disease, and Hopkinson suddenly became a minority, thousands of miles from home.

Development can be described as an orderly alienation. In mammals, zygotes divide and subsets of cells become functionally specialized into, say, neurons or liver cells. Following the discovery of DNA as the genetic material in the 1950s, a question arose: did dividing cells retain all genes from the zygote, or were genes lost as it specialized? British embryologist John Gurdon addressed this question in a series of experiments in the 60s using frogs. Gurdon transplanted nuclei from varyingly differentiated cells into oocytes stripped of their genetic material to see if a new frog was made. He found the more differentiated a cell was, the lower the chance of success, but the successes confirmed that no genetic material was lost. Meanwhile, Canadian biologists Ernest McCulloch and James Till were transplanting bone marrow to treat irradiated mice when they noticed it caused lumps in the mices spleens, and the number of lumps correlated with the cellular dosage. Their lab subsequently demonstrated that each lump was a clonal colony from a single donor cell, and a subset of those cells was self-renewing and could form further colonies of any blood cell type. They had discovered hematopoietic stem cells. In 1981 the first embryonic stem cells (ESCs) from mice were successfully propagated in culture by British biologist Martin Evans, winning him the Nobel Prize in 2007. This breakthrough allowed biologists to alter genes in ESCs, then use Gurdons technique to create transgenic mice with that alteration in every cellcreating the first animal models of disease.

In 1982, one year after Evans discovery, Hopkinson graduated with honors from York University. She worked in the arts, as a library clerk, government culture research officer, and grants officer for the Toronto Arts Council, but wouldnt begin publishing her own fiction until she was 34. [I had been] politicized by feminist and Caribbean literature into valuing writing that spoke of particular cultural experiences of living under colonialism/patriarchy, and also of writing in ones own vernacular speech, Hopkinson said. In other words, I had models for strong fiction, and I knew intimately the body of work to which I would be responding. Then I discovered that Delany was a black man, which opened up a space for me in SF/F that I hadnt known I needed. She sought out more science fiction by black authors and found Butler, Charles Saunders, and Steven Barnes. Then the famous feminist science fiction author and editor Judy Merril offered an evening course in writing science fiction through a Toronto college, Hopkinson said. The course never ran, but it prompted me to write my first adult attempt at a science fiction story. Judy met once with the handful of us she would have accepted into the course and showed us how to run our own writing workshop without her. Hopkinsons dream of attending Clarion came true in 1995, with Delany as an instructor. Her early short stories channeled her love of myth and folklore, and her first book, written in Caribbean dialect, married Caribbean myth to the science fictional trappings of black market organ harvesting. Brown Girl in the Ring (1998) follows a young single mother as shes torn between her ancestral culture and modern life in a post-economic collapse Toronto. It won the Aspect and Locus Awards for Best First Novel, and Hopkinson was awarded the John W. Campbell Award for Best New Writer.

In 1996, Dolly the Sheep was created using Gurdons technique to determine if mammalian cells also could revert to more a more primitive, pluripotent state. Widespread animal cloning attempts soon followed, (something Hopkinson used as a science fictional element in Brown Girl) but it was inefficient, and often produced abnormal animals. Ideas of human cloning captured the public imagination as stem cell research exploded onto the scene. One ready source for human ESC (hESC) materials was from embryos which would otherwise be destroyed following in vitro fertilization (IVF) but the U.S. passed the Dickey-Wicker Amendment prohibited federal funding of research that destroyed such embryos. Nevertheless, in 1998 Wisconsin researcher James Thomson, using private funding, successfully isolated and cultured hESCs. Soon after, researchers around the world figured out how to nudge cells down different lineages, with ideas that transplant rejection and genetic disease would soon become things of the past, sliding neatly into the hole that the failure of genetic engineering techniques had left behind. But another blow to the stem cell research community came in 2001, when President Bushs stem cell ban limited research in the U.S. to nineteen existing cell lines.

In the late 1990s, another piece of technology capturing the public imagination was the internet, which promised to bring the world together in unprecedented ways. One such way was through private listservs, the kind used by writer and academic Alondra Nelson to create a space for students and artists to explore Afrofuturist ideas about technology, space, freedom, culture and art with science fiction at the center. It was wonderful, Hopkinson said. It gave me a place to talk and debate with like-minded people about the conjunction of blackness and science fiction without being shouted down by white men or having to teach Racism 101. Connections create communities, which in turn create movements, and in 1999, Delanys essay, Racism and Science Fiction, prompted a call for more meaningful discussions around race in the SF community. In response, Hopkinson became a co-founder of the Carl Brandon society, which works to increase awareness and representation of people of color in the community.

Hopkinsons second novel, Robber, was a breakthrough success and was nominated for Hugo, Nebula, and Tiptree Awards. She would also release Skin Folk (2001), a collection of stories in which mythical figures of West African and Afro-Caribbean culture walk among us, which would win the World Fantasy Award and was selected as one ofThe New York Times Best Books of the Year. Hopkinson also obtained masters degree in fiction writing (which helped alleviate U.S. border hassles when traveling for speaking engagements) during which she wrote The Salt Roads (2003). I knew it would take a level of research, focus and concentration I was struggling to maintain, Hopkinson said. I figured it would help to have a mentor to coach me through it. That turned out to be James Morrow, and he did so admirably. Roads is a masterful work of slipstream literary fantasy that follows the lives of women scattered through time, bound together by the salt uniting all black life. It was nominated for a Nebula and won the Gaylactic Spectrum Award. Hopkinson also edited anthologies centering around different cultures and perspectives, including Whispers from the Cotton Tree Root: Caribbean Fabulist Fiction (2000), Mojo: Conjure Stories (2003), and So Long, Been Dreaming: Postcolonial Science Fiction & Fantasy (2004). She also came out with the award-winning novelThe New Moons Arms in 2007, in which a peri-menopausal woman in a fictional Caribbean town is confronted by her past and the changes she must make to keep her family in her life.

While the stem cell ban hamstrung hESC work, Gurdons research facilitated yet another scientific breakthrough. Researchers began untangling how gene expression changed as stem cells differentiated, and in 2006, Shinya Yamanaka of Kyoto University reported the successful creation of mouse stem cells from differentiated cells. Using a list of 24 pluripotency-associated genes, Yamanaka systematically tested different gene combinations on terminally differentiated cells. He found four genesthereafter known as Yamanaka factorsthat could turn them into induced-pluripotent stem cells (iPSCs), and he and Gurdon would share a 2012 Nobel prize. In 2009, President Obama lifted restrictions on hESC research, and the first clinical trial involving products made using stem cells happened that year. The first human trials using hESCs to treat spinal injuries happened in 2014, and the first iPSC clinical trials for blindness began this past December.

Hopkinson, too, encountered complications and delays at points in her career. For years, Hopkinson suffered escalating symptoms from fibromyalgia, a chronic disease that runs in her family, which interfered with her writing, causing Hopkinson and her partner to struggle with poverty and homelessness. But in 2011, Hopkinson applied to become a professor of Creative Writing at the University of California, Riverside. It seemed in many ways tailor-made for me, Hopkinson said. They specifically wanted a science fiction writer (unheard of in North American Creative Writing departments); they wanted someone with expertise working with a diverse range of people; they were willing to hire someone without a PhD, if their publications were sufficient; they were offering the security of tenure. She got the job, and thanks to a steady paycheck and the benefits of the mild California climate, she got back to writing. Her YA novel, The Chaos (2012), coming-of-age novelSister Mine (2013), and another short story collection, Falling in Love with Hominids (2015) soon followed. Her recent work includes House of Whispers (2018-present), a series in DC Comics Sandman Universe, the final collected volume of which is due out this June. Hopkinson also received an honorary doctorate in 2016 from Anglia Ruskin University in the U.K., and was Guest of Honor at 2017 Worldcon, a year in which women and people of color dominated the historically white, male ballot.

While the Yamanaka factors meant that iPSCs became a standard lab technique, iPSCs are not identical to hESCs. Fascinatingly, two of these factors act together to maintain the silencing of large swaths of DNA. Back in the 1980s, researchers discovered that some regions of DNA are modified by small methyl groups, which can be passed down through cell division. Different cell types have different DNA methylation patterns, and their distribution is far from random; they accumulate in the promoter regions just upstream of genes where their on/off switches are, and the greater the number of methyl groups, the lesser the genes expression. Furthermore, epigenetic modifications, like methylation, can be laid down by our environments (via diet, or stress) which can also be passed down through generations. Even some diseases, like fibromyalgia, have recently been implicated as such an epigenetic disease. Turns out that the long-standing biological paradigm that rejected Lamarck also missed the bigger picture: Nature is, in fact, intimately informed by nurture and environment.

In the past 150 years, we have seen ideas of community grow and expand as the world became more connected, so that they now encompass the globe. The histories of science fiction and biology are full of stories of pioneers opening new doorsbe they doors of greater representation or greater understanding, or bothand others following. If evolution has taught us anything, its that nature abhors a monoculture, and the universe tends towards diversification; healthy communities are ones which understand that we are not apart from the world, but of it, and that diversity of types, be they cells or perspectives, is a strength.

Kelly Lagor is a scientist by day and a science fiction writer by night. Her work has appeared at Tor.com and other places, and you can find her tweeting about all kinds of nonsense @klagor

See the original post:
On the Origins of Modern Biology and the Fantastic: Part 19 Nalo Hopkinson and Stem Cell Research - tor.com

Stem Cell Alopecia Treatment Market Growth Analysis by Size, Top Companies, Supply Demand, Trends, Demand, Overview and Forecast to 2026 – Cole of…

New Jersey, United States, The Stem Cell Alopecia Treatment Market report examines the market situation and prospects and represents the size of the Stem Cell Alopecia Treatment market (value and volume) and the share by company, type, application and region. The general trends and opportunities of Stem Cell Alopecia Treatment are also taken into account when examining the Stem Cell Alopecia Treatment industry. Stem Cell Alopecia Treatment The market report focuses on the following section: Analysis of the Stem Cell Alopecia Treatment industry by transfer into different segments; the main types of products that fall within the scope of the report.

This Stem Cell Alopecia Treatment market report is a complete analysis of the Stem Cell Alopecia Treatment market based on an in-depth primary and secondary analysis. The scope of the Stem Cell Alopecia Treatment market report includes global and regional sales, product consumption in terms of volume and value. The Stem Cell Alopecia Treatment market report contains an estimate of revenue, CAGR and total revenue. The knowledge gathered in world trade Stem Cell Alopecia Treatment is presented in figures, tables, pie charts and graphics.

Get | Research Sample Copy By Using Corporate Mail ID @https://www.verifiedmarketresearch.com/download-sample/?rid=15102&utm_source=COD&utm_medium=007

Top 10 Companies in the Global Stem Cell Alopecia Treatment Market Research Report:

Global Stem Cell Alopecia Treatment Market: Drivers and Restrains

The research report included analysis of various factors that increase market growth. These are trends, restrictions and drivers that change the market positively or negatively. This section also contains information on various segments and applications that may affect the market in the future. Detailed information is based on current trends and historical milestones. This section also includes an analysis of sales volume on the Stem Cell Alopecia Treatment market and for each type from 2015 to 2026. This section mentions sales volume by region from 2015 to 2026. The price analysis is included in the report Type of year 2015 to 2026, manufacturer from 2015 to 2020, region from 2015 to 2020 and total price from 2015 to 2026.

An in-depth assessment of the restrictions contained in the report describes the contrast to the drivers and leaves room for strategic planning. The factors that overshadow the growth of the market are essential as they can be understood to design different phrases to take advantage of the lucrative opportunities that the growing Stem Cell Alopecia Treatment market offers. In addition, information on the opinions of market experts was used to better understand the market.

Global Stem Cell Alopecia Treatment Market: Segment Analysis

The research report contains certain segments such as application and product type. Each type provides revenue information for the 2015-2026 forecast period. The application segment also provides volume revenue and revenue for the 2015-2026 forecast period. Understanding the segments identifies the importance of the various factors that support Stem Cell Alopecia Treatment market growth.

Global Stem Cell Alopecia Treatment Market: Regional Analysis

The research report includes a detailed study of the regions of North America, Europe, Asia Pacific, Latin America, the Middle East and Africa. The Stem Cell Alopecia Treatment report was compiled after various factors determining regional growth, such as the economic, environmental, social, technological and political status of the region concerned, were observed and examined. Analysts examined sales, sales, and manufacturer data for each region. This section analyzes sales and volume by region for the forecast period from 2015 to 2026. These analyzes help the reader understand the potential value of investments in a particular region.

Global Stem Cell Alopecia Treatment Market: Competitive Landscape

This section of the report lists various major manufacturers in the market. It helps the reader understand the strategies and collaborations that players focus on to fight competition in the market. The full report provides a significant microscopic overview of the Stem Cell Alopecia Treatment market. Readers can identify manufacturers footprints by knowing manufacturers global earnings, manufacturers world market prices, and manufacturers sales for the 2015-2019 forecast period.

Ask For Discounts, Click Here @ https://www.verifiedmarketresearch.com/ask-for-discount/?rid=15102&utm_source=COD&utm_medium=007

Table of Content

1 Introduction of Stem Cell Alopecia Treatment Market

1.1 Overview of the Market1.2 Scope of Report1.3 Assumptions

2 Executive Summary

3 Research Methodology of Verified Market Research

3.1 Data Mining3.2 Validation3.3 Primary Interviews3.4 List of Data Sources

4 Stem Cell Alopecia Treatment Market Outlook

4.1 Overview4.2 Market Dynamics4.2.1 Drivers4.2.2 Restraints4.2.3 Opportunities4.3 Porters Five Force Model4.4 Value Chain Analysis

5 Stem Cell Alopecia Treatment Market, By Deployment Model

5.1 Overview

6 Stem Cell Alopecia Treatment Market, By Solution

6.1 Overview

7 Stem Cell Alopecia Treatment Market, By Vertical

7.1 Overview

8 Stem Cell Alopecia Treatment Market, By Geography

8.1 Overview8.2 North America8.2.1 U.S.8.2.2 Canada8.2.3 Mexico8.3 Europe8.3.1 Germany8.3.2 U.K.8.3.3 France8.3.4 Rest of Europe8.4 Asia Pacific8.4.1 China8.4.2 Japan8.4.3 India8.4.4 Rest of Asia Pacific8.5 Rest of the World8.5.1 Latin America8.5.2 Middle East

9 Stem Cell Alopecia Treatment Market Competitive Landscape

9.1 Overview9.2 Company Market Ranking9.3 Key Development Strategies

10 Company Profiles

10.1.1 Overview10.1.2 Financial Performance10.1.3 Product Outlook10.1.4 Key Developments

11 Appendix

11.1 Related Research

Customized Research Report Using Corporate Email Id @https://www.verifiedmarketresearch.com/product/stem-cell-alopecia-treatment-market/?utm_source=COD&utm_medium=007

About us:

Verified Market Research is a leading Global Research and Consulting firm servicing over 5000+ customers. Verified Market Research provides advanced analytical research solutions while offering information enriched research studies. We offer insight into strategic and growth analyses, Data necessary to achieve corporate goals and critical revenue decisions.

Our 250 Analysts and SMEs offer a high level of expertise in data collection and governance use industrial techniques to collect and analyse data on more than 15,000 high impact and niche markets. Our analysts are trained to combine modern data collection techniques, superior research methodology, expertise and years of collective experience to produce informative and accurate research.

We study 14+ categories from Semiconductor & Electronics, Chemicals, Advanced Materials, Aerospace & Defence, Energy & Power, Healthcare, Pharmaceuticals, Automotive & Transportation, Information & Communication Technology, Software & Services, Information Security, Mining, Minerals & Metals, Building & construction, Agriculture industry and Medical Devices from over 100 countries.

Contact us:

Mr. Edwyne Fernandes

US: +1 (650)-781-4080UK: +44 (203)-411-9686APAC: +91 (902)-863-5784US Toll Free: +1 (800)-7821768

Email: [emailprotected]

The rest is here:
Stem Cell Alopecia Treatment Market Growth Analysis by Size, Top Companies, Supply Demand, Trends, Demand, Overview and Forecast to 2026 - Cole of...

Monocyte-derived multipotent cell delivered programmed therapeutics to reverse idiopathic pulmonary fibrosis – Science Advances

INTRODUCTION

Idiopathic pulmonary fibrosis (IPF) is a rapidly progressive and fatal interstitial pulmonary disease with a dismal median survival time of just 3 years after diagnosis (1, 2). To date, the IPF therapies depend on blocking myofibroblast activation to inhibit collagen I deposition (3, 4). However, the clinical data showed that these therapies remained far from achieving IPF revision. The main reason is that the IPF therapeutics lack an effectively targeted carrier or ignore some of the other risk factors such as the instability and tolerability of type II alveolar epithelial cell (AEC II) (5, 6). The AEC II, which is considered as injured AEC II (7, 8) in the IPF tissues, releases excessive amounts of reactive oxygen species (ROS) that initiate an antifibrinolytic coagulation cascade and promote the overexpression of connective tissue growth factor (CTGF) to provoke myofibroblast overactivation and extracellular matrix (ECM) development and then destroy the lung architecture (911). This situation has inspired us to propose that the combination of modulating superoxide in injured AEC II and antimyofibroblast activation as weeding and uprooting strategy will be a potential therapeutic strategy for synergistic antifibrosis. Furthermore, another limitation is that current therapies are rarely distributed in the lungs, which cannot achieve full therapeutic effect for treating IPF (12). Thus, the development of an effective lung-targeting drug delivery carrier is highly desirable for IPF therapy.

Recently, local preferred therapeutic agents generated using endogenous cells have served as a strong and promising delivery platform for targeting in situ, achieving considerable progress in several diseases (1317). In the inflammatory phase of IPF, precursor circulating monocytes (PCMs) have been found to undergo notable proliferation (18). PCMs and injured AEC II release chemotactic factors that specifically recruit chemokine receptorpositive (CR+) cells including monocyte-derived multipotent cell (MOMC) and guide the MOMC to migrate to injured lung tissues through specific binding to chemokine receptors on cell membrane (19, 20). Furthermore, in addition to this migration characteristic, these MOMCs, which originate from hematopoietic stem cells in the bone marrow, still have multipotency to differentiate into a variety of functional cells, including AEC II and endothelial cell (21, 22), which demonstrates that MOMC has the potential to participate in reestablishing lung functions (23). In addition, chronic hypoxic exposure induces the recruitment of MOMC to the pulmonary circulation, and the cell contributes to improving lung functions by producing angiogenic factors (24). It has been reported that monocytes from patients with IPF also show preconditioned prorepair features (25). In general, MOMC as a precise lung-targeting delivery platform will exhibit encouraging therapeutic effects, leading to the repair or regeneration of injured AEC II for IPF treatment.

In this study, we constructed the programmed therapeutics composed of surface-engineered nanoparticles (PER NPs) loading dual drugs adhered to MOMC (named MOMC/PER) to solve the issues in IPF therapy by improving drug accumulation in injured lung sites and completely destroying the fibrotic signaling network in IPF (Fig. 1). The MOMC/PER delivery platform realized efficacy through programmed modules, which consisted of a homing moiety, responsive release moiety, and retargeting moiety. (i) The homing moiety is the native ability of MOMC/PER to migrate to injured lungs due to the homing characteristic of MOMC. (ii) The responsive release moiety of MOMC/PER is activated by matrix metalloproteinase-2 (MMP-2) overexpression in IPF tissues, resulting in pathology-responsive release of PER NPs with exposed cyclic RGDfc (Arg-Gly-Asp) [c(RGDfc)] from the MOMC. (iii) The retargeting moiety is that exposed c(RGDfc) on PER NPs can anchor to injured AEC II via an interaction between v6 and c(RGDfc) (26), allowing the cytoplasm of the injured AEC II internalize PER NPs. Subsequently, astaxanthin (AST) and trametinib (TRA) are released from PER NPs to achieve a weeding and uprooting therapeutic effect. In general, the sustained injury of epithelial cells and highly heterogeneous myofibroblasts is considered as the most critical variable in achieving complete IPF reversion (27). To validate the above hypothesis, in this study, AST was chosen as an antioxidant by neutralizing superoxide to repair injured AEC II (28), and TRA suppressed the activation of myofibroblast by inhibiting CTGF production for IPF therapy (29). MOMC also participates in treating IPF by repairing injured AEC II to promote regeneration of IPF lungs (21). Overall, MOMC/PER, which mimics the features of chimeric antigen receptor T cell immunotherapy, is a precise lung-targeting platform to reverse IPF by improving drug accumulation due to the outstanding homing ability of MOMC to injured lung sites, and the destruction of the fibrotic signaling network by inhibiting the activation of myofibroblast and repairing injured AEC II to promote the damaged lungs regeneration.

(A) Bioconjugated MOMC/PER was prepared by incubating PER NPs with MOMC. (B) MOMC/PER has multifunctional moieties including a homing moiety, responsive release moiety, and retargeting moiety to reverse IPF. Then, a weeding and uprooting strategy contributes to IPF reversion. (C) Schematic illustration of MOMC/PER for improved drugs accumulation and antifibrotic effect in IPF lung microenvironment.

The quantities of MOMC in serum and lung tissues were significantly increased in IPF mice compared with normal mice (Fig. 2A). The proliferation of MOMC was positively related to IPF progression, which might be because increasing numbers of MOMC would be recruited from the bone marrow to the lesion sites when IPF occurred (30). Motivated by the fact that MOMC has a homing ability, we considered MOMC to be a potential delivery carrier to improve delivery efficiency in IPF treatment under pathological conditions.

(A) The proliferation of Nanog+ cells in serum and lung tissues by ELISA assay. (B) The MOMC phenotypes. The level of TGF- (C) and hydroxyproline (D) in IPF lung tissues, respectively. (E) The level of TGF-/Smad in vitro. (F) Schematic showing the preparation of MOMC/PER. (G) Schematic showing the adhesion of PER NPs to MOMC. (H) SEM images of MOMC and MOMC/PER-DiI. (I) Fluorescent signals of MOMC and PER-DiI NPs by CLSM. (J) The adhesion between MOMC and PER-DiI NPs by flow cytometry. (K) In vitro migration model. The migration ability of MOMC and MOMC/PER in CXCL 12 (L) and CCL 19 (M), respectively. (N) Schematic showing sensitive release of MOMC/PER-DiI triggered by MMP-2. (O) Characterizations by TEM. MOMC is the triangle, and PER-DiI NPs are the arrows. (P) Fluorescent images of MOMC and released PER-DiI NPs by CLSM. (Q) The flow cytometry showed responsive release. (R) Schematic showing the retarget ability of released PER NPs. (S) Characterization of retargeting ability by TEM. (T) The fluorescent images by CLSM. (U) Cellular uptake in A549 by flow cytometry (n = 3). Statistical significance was calculated via one-way analysis of variance (ANOVA).

We first isolated MOMC from the peripheral blood of C57BL/6J male mice of IPF. The morphologies of the MOMC were fusiform (fig. S1). To identify the phenotypes of MOMC isolated from IPF mice, we first investigated the presence of specific markers for MOMC by immunofluorescence staining. The results showed that MOMC expressed CD11b and smooth muscle actin (-SMA) (Fig. 2B), which was consistent with the literature (24). In addition, the MOMC also expressed the stem cell markers CD14 and Nanog protein and the injured AEC IIs marker pro-surfactant protein C (SPC), as shown in Fig. 2B. These results indicated that MOMC was pluripotent cells with stem cell and epithelial celllike properties. It has been reported that MOMC was recruited to damaged lung areas and participated in recovering injured lung normalization through growth factor release to repair injured AEC II (30). In addition, to inspect the potential risk of injecting MOMC into mice, we further investigated the feasibility of using isolated MOMC as a delivery carrier, including measuring the levels of transforming growth factor (TGF-) and hydroxyproline, which are closely related to the development of IPF in vivo. The results displayed approximately onefold reduction in TGF- and hydroxyproline levels in IPF mice treated with MOMC compared with untreated bleomycin (BLM)induced mice, and these indexes were barely changed in normal mice, indicating that MOMC would not induce the occurrence of IPF and partly relieved established IPF (Fig. 2, C and D).

We next prepared PER NPs that contained two target peptides named peptide E5 and c(RGDfc). The poly(lactide-co-glycolide)-block-poly(ethylene glycol) methyl ether maleimide (PLGA-PEG-Mal) and PLGA-PEG-c(RGDfc) (mass ratio, 10:1) were self-assembled by noncovalent interactions of amphiphilic PLGA-PEG copolymer into nanoparticles (31), and then, peptide E5 was bound on the NPs by the Michael reaction (fig. S2A). As determined by 1H nuclear magnetic resonance spectroscopy (fig. S2B) and SDSpolyacrylamide gel electrophoresis (SDS-PAGE) (fig. S2C), we successfully prepared PER NPs, and the grafting rate of peptide E5 in the PER NPs was 43.7%. The PER NPs showed particle sizes of approximately 110 10.39 nm and the zeta potential of 23.37 mV (fig. S2, D and E). In addition, AST and TRA were encapsulated into PER NPs (fig. S2F). The drug loading content of the PER NPs was 1.98 weight % (wt %) for AST and 2.83 wt % for TRA. The sustained release of the loaded AST was 49.5 wt %, and the pH-dependent release of the loaded TRA (weak alkalinity) was 79.6 wt %, which were obtained at pH 5.0 within 72 hours (fig. S2G). Then, we investigated the capacity of MOMC to differentiate into myofibroblast after treatment with PER NPs in vitro. As shown in Fig. 2E, the expression of TGF-/small mother against decapentaplegic (TGF-/Smad), which is molecule in the crucial pathway for myofibroblast activation, was decreased, suggesting that the PER NPs could inhibit MOMC differentiation. The possible reason for the inhibition was that the PER NPs partly covered the TGF- receptor on the MOMC and reduced exogenous TGF- stimulation within 8 hours, and then, the PER NPs could be gradually internalized. The released drugs could reduce TGF- expression of MOMC after 8 hours (fig. S3, A and B).

We next constructed MOMC/PER as a delivery platform/therapeutic carrier (Fig. 2F), and PER NPs loaded with both drugs could specifically adhere to MOMC through the interaction between peptide E5 of the PER NPs and the CXCR4 on the MOMC by a temperature-dependent manner (32, 33). The formation of MOMC/PER was positively correlated with the incubation time within 2 hours (fig. S3, A and B). Moreover, the PER NPs could specifically stick to the surface of the MOMC without internalization by the MOMC within 8 hours (Fig. 2G). The reasons may be that peptide E5 conjugated on the surface of the PER NPs is a long-chain peptide that limits internalization into the MOMC and that CXCR4 is not an endocytic receptor (34). The MOMC/PER had a loading capacity of 4.75 g of TRA and 1.5 g of AST/1 105 cells (fig. S3, C and D). In addition, MOMC cell viability was above 80% with different concentrations of PER NPs and different incubation times (fig. S4, A and B).

To investigate the adhesion of MOMC and PER NPs, the 1,1-dioctadecyl-3,3,3,3-tetramethylindocarbocyanine perchlorate (DiI) was loaded into blank PER NPs (PER-DiI NPs) to evaluate adhesion behavior. After incubating with MOMC and PER-DiI NPs for 2 hours, the morphologies of the MOMC/PER-DiI were confirmed by scanning electron microscopy (SEM) (Fig. 2H) and confocal laser scanning microscopy (CLSM) (Fig. 2I). Flow cytometry detection also indicated the formation of MOMC/PER-DiI in that the MOMC labeling green and PER-DiI NPs were collected in the double-positive quadrant (Fig. 2J).

Sequentially, migration via the interaction between a receptor and ligand is the vital characteristic that needs to be retained by MOMC/PER to realize efficient delivery (Fig. 2K). The migratory capability of MOMC/PER was detected by a Transwell invasion assay. The results indicated that the migratory ability of the MOMC/PER was unaffected by PER NPs adhering to the surface of MOMC (Fig. 2, L and M) at all studied concentrations (fig. S4, C and D).

To establish the retargeting ability of PER NPs, MMP-2 overexpressed in IPF tissues was used as an activating trigger to release PER NPs from MOMC/PER. As depicted in Fig. 2N, the separation of PER-DiI NPs from MOMC was well evidenced by transmission electron microscopy (TEM) and CLSM (Fig. 2, O and P) and flow cytometry (Fig. 2Q). We also detected the phenomenon by SDS-PAGE and particle sizes changes (fig. S4, E and F). After PER NPs were released from MOMC/PER, the exposed peptide c(RGDfc) of the PR NPs could retarget v6, which is overexpressed on the surface of injured AEC II (Fig. 2R) (35). Then, we investigated the capacity of injured AEC II to uptake PLGA-PEG-c(RGDfc)coumarin 6 (PR-C6) compared with free C6 and PLGA-PEGC6 (PP-C6) by TEM and CLSM. The internalization of PR-C6 was better than other forms (C6 and PP-C6) (Fig. 2, S to U). In addition, PR-C6 also underwent lysosomal escape (fig. S4G).

The homing ability of MOMC/PER was investigated in IPF models in vivo (Fig. 3A). We first examined the lung accumulation of 1,1-dioctadecyl-3,3,3,3-tetramethylindotricarbocyanine iodide (DiR)loaded into blank PER NPs (PER-DiR NPs) adhere to MOMC (MOMC/PER-DiR) after intravenous administration. The DiR fluorescence accumulated in the lungs of the MOMC/PER-DiR group, which indicated that compared with MOMC-loading DiR (MOMC-DiR) and free-DiR, MOMC/PER-DiR had a superior ability to target IPF lungs (Fig. 3B). Then, we quantitatively analyzed the drug distribution in the tissues of each organ. The DiR fluorescence intensity in the lungs was 3.5- and 0.5-fold greater than that in the liver in the MOMC/PER-DiR and MOMC-DiR groups, respectively. In addition, there was little accumulation of free DiR in the lungs than that in the liver (Fig. 3C). MOMC loading of DiR could improve DiR accumulation in IPF lungs due to the homing ability of the MOMC.However, the accumulation of MOMC-DiR was weaker than that of MOMC/PER-DiR. This may be because the free dye carried by the MOMC was limited compared with that carried by the PER NPs, which suggested that MOMC/PER could solve the limitation of conventional drug loading of cells. In addition, we further evaluated the homing capacity of MOMC/PER, the responsive release ability of MOMC/PER mediated by MMP-2, the released PER NPs with exposed c(RGDfc), and the retargeting to injured AEC II by immunofluorescence staining. The DiI was chose to a tracer agent, labeling the PER-DiI NPs with red fluorescence, and then, the PER-DiI NPs adhered to MOMC to form MOMC/PER-DiI. Nanog and SPC, which represented MOMC and injured AEC II, respectively, were labeled in green fluorescence. Then, the MOMC/PER-DiI was administered to IPF mice by intravenous injection. As shown in Fig. 3D, the PER-DiI NPs labeled in red overlapped with the MOMC marked in green, generating a merged yellow signal, which revealed that the MOMC/PER-DiI notably accumulated in the lungs of IPF mice in stage 1 (homing to lungs). In stage 2 (releasing PER-DiI NPs), the PER-DiI NPs labeled in red were separated from the MOMC labeled in green, indicating that the PER-DiI NPs were released from the MOMC membrane surface and exposed c(RGDfc) at fibrotic foci as a result of the overexpression of MMP-2 in the IPF microenvironment. Then, the PER-DiI NPs labeled in red overlapped with injured AEC IISPC+ labeled in green, implying that the PER-DiI NPs retargeted to injured AEC II through the interaction between the exposed c(RGDfc) ligand and the v6 receptor on the surface of the injured AEC II in stage 3 (retarget injured AEC II) (Fig. 3D). Collectively, these results showed that MOMC/PER-DiI had the native ability to home to damaged lungs and then were activated by programmed procedures, confirming that MOMC could function as a vehicle to deliver PER NPs to injured lungs.

(A) Schematic of the targeting performance of MOMC/PER in the blood circulation to IPF lungs. (B) In vivo fluorescence images of IPF mice intravenous injection with MOMC-DiR, MOMC/PER-DiR, and DiR (n = 3). (C) Quantification of the in vivo retention profile (n = 3). (D) The different stages of MOMC/PER-DiI. (E) The whole lungs were imaged and investigated after 28 days. Lung morphologies (i) [Photo credit (i): Xin Chang, China Pharmaceutical University], H&E staining (ii), and Masson staining (iii). The morphologies of mitochondria by TEM (iv). The levels of TGF- (F), IL-1 (G), and IL-4 (H) by ELISA assay (n = 5). The levels of lymphocytes (I), white blood cells (J), and neutrophils (K) in whole blood (n = 5). The levels of GSH (L) and SOD (M), respectively (n = 5). (N) The expression of SPC. (O) Survival rate curves (n = 10). Statistical significance was calculated via one-way ANOVA.

To confirm the curative effect of MOMC/PER, we investigated lung morphologies after the administration of MOMC/PER or other treatments. As showed in Fig. 3E, MOMC/PER could greatly relieve IPF according to hematoxylin and eosin (H&E) and Masson staining. Images of lung morphologies showed obvious normalization after treatment with MOMC or MOMC/PER compared with no treatment (Fig. 3E, i). H&E staining showed that lung tissues in the MOMC/PER group were not destroyed and that the alveolar sizes were same as normal lung tissues (Fig. 3E, ii). In addition, compared with no treatment, MOMC also partly protected the lung architecture; however, there was a gap between the MOMC/PER and normal groups. Similarly, Masson staining also showed that the MOMC/PER group exhibited an excellent reduction in collagen I deposition (Fig. 3E, iii). IPF is also induced by mitochondrial oxidative stress in injured AEC II. Hence, we examined the capability of MOMC/PER to repair injured AEC II by maintaining mitochondrial morphologies (Fig. 3E, iv). The morphologies of mitochondria were close to normal in the MOMC/PER group compared with the MOMC group and BLM group, suggesting that MOMC/PER could repair injured AEC II to maintain normal lungs by improving mitochondrial function. Furthermore, we tested the expression of proinflammatory cytokines [TGF-, interleukin-1 (IL-1), and IL-4], which play major roles in excessive ECM formation during IPF progression. As shown in Fig. 3 (F to H), the expression of TGF- in the MOMC/PER treatment group was nearly threefold lower than that in the BLM group, and the expression of IL-1 and IL-4 also decreased by nearly 0.5- and 1-fold, respectively, in the MOMC/PER group compared with the BLM group, suggesting that MOMC/PER could block IPF progression by inhibiting the secretion of proinflammatory cytokines. In addition, the formulations of MOMC and MOMC/PER showed well biocompatibility in a hemolysis test (fig. S5). In addition, inflammatory cells were quantified in whole blood in these groups after treatment. Compared with the BLM group, the MOMC/PER group showed inhibited inflammatory cell proliferation (Fig. 3, I to K), which indicated that MOMC/PER had the ability to alleviate IPF progression in the inflammatory phase. In addition, the results implied that MOMC had a certain ability to inhibit the proliferation of inflammatory cells. Next, glutathione (GSH) and superoxide dismutase (SOD), which are significant inhibitors of ROS, were used to balance the ROS content of injured AEC II. Compared with no treatment, treatment with MOMC/PER increased the GSH level nearly onefold (Fig. 3L), and MOMC also enhanced the GSH level. Similarly, MOMC/PER increased the SOD level to a certain extent in lung tissues (Fig. 3M). We further explored the repair mechanism for injured AEC II in IPF lungs treated with MOMC or MOMC/PER. The expression of SPC was markedly increased in the MOMC/PER group compared with the BLM group; there was also an augmentation in the expression of SPC in the MOMC group, which showed that MOMC/PER could up-regulate AEC II proliferation or recover injured AEC II to normalize the lungs in IPF and demonstrated that MOMC/PER could promote IPF lungs regeneration (Fig. 3N). The survival time of the MOMC/PER group exceeded 60 days, which was longer than the survival time of the BLM group (Fig. 3O), and the MOMC/PER group did not exhibit any changes in body weight (fig. S6).

To investigate the targeting ability of PER NPs through reprogramming to form MOMC/PER in the blood circulation, we conducted the following experiments. The E5-mediated targeting ability of PER-DiI NPs was first evaluated in IPF mice (Fig. 4A). CLSM showed the adhesion of PER-DiI NPs to the surface of MOMC (Fig. 4B, bottom). The confocal images produced the same result as Fig. 2I. Furthermore, PER-DiI NPs were administered to IPF mice model by intravenous injection. The results demonstrated that the PER-DiI NPs adhered to the surface of MOMC (Fig. 4B, middle). More detailed results revealed that the PER-DiI NPs could bind to the MOMC surface and reprogram the MOMC to form MOMC/PER in the peripheral blood by SEM (Fig. 4B, top). In addition, immunofluorescence staining confirmed that the PER-DiI NPs homed to IPF lungs and accumulated in the injured AEC II area after intravenous injection (Fig. 4C). In addition, Nanog-labeled MOMC (green fluorescence) accumulated in higher numbers in IPF lungs than normal lungs, which was similar to previous results (Fig. 2I). We also investigated the targeting capacity of PER-DiR NPs at different time points by in vivo imaging system following intravenous injection, and PLGA-PEG-DiR (PP-DiR NPs), PLGA-PEG-c(RGDfc)DiR (PR-DiR NPs), and PLGA-PEG-E5-DiR (PE-DiR NPs) were used as controls. The PP-DiR NPs and PR-DiR NPs were mainly found in the liver, while the PER-DiR NPs and PE-DiR NPs mainly accumulated in IPF lungs (Fig. 4D and fig. S7A). The accumulation of the PER-DiR NPs in the lungs peaked at 8 hours, while the lung accumulation of the PE-DiR NPs quickly decreased. The primary reason may be that the PE NPs were delivered to the lungs via MOMC; however, they could not anchor on injured AEC II because they lacked c(RGDfc) and were therefore more rapidly cleared from the circulation than the PER-DiR NPs. Compared with the PE-DiR NPs, the PER-DiR NPs accumulated in IPF lungs for a long time (more than 8 hours), which is important for treating lung disease. A quantitative region of interest (ROI) analysis of PER-DiR NPs accumulation was performed by detecting DiR signal variation in the lungs and other organs (Fig. 4E). Moreover, we evaluated PER-DiI NPs behavior in lung tissues after administration at different times (Fig. 4F). After administration at 0.5 hours, increasing levels of overlapping yellow fluorescence in lung blood vessels were observed for PER-DiI NPs labeled in red and MOMC marked in green, indicating that the PER-DiI NPs arrived at IPF lung tissues through reprogramming to form MOMC/PER-DiI in the blood circulation. Then, the red and green signals were separated at the time point of 2 hours, indicating that the PER-DiI NPs were released from the reprogrammed MOMC/PER-DiI due to the overexpression of MMP-2 in the IPF mice. Furthermore, the released PER-DiI NPs showed a wide distribution in the lung tissues at 4 hours after intravenous injection, which is powerful for treating diseases. These data demonstrated that PER NPs could target IPF lungs by means of attaching to circulating MOMC quickly and could accumulate in lung tissues for a long time to achieve therapeutic efficacy.

(A) Schematic of PER NPs circulation in vivo, reprogramming of MOMC/PER, and recruitment to IPF tissue. (B) The targeting ability of PER-DiI NPs. (C) The accumulation of PER-DiI NPs in normal and IPF lungs. (D) Fluorescence IVIS imaging (n = 3). (E) Ex vivo fluorescence imaging and quantification of major organs (n = 3). (F) The accumulation PER-DiI NPs in the lungs at different times. Lung function indexes of GSH (G), SOD (H), and MDA (I). TGF- (J), IL-1 (K), and IL-4 (L) by ELISA assay (n = 5). (M) Proliferation of fibroblasts. (N) Expression of collagen I. Statistical significance was calculated via one-way ANOVA.

We further investigated the antifibrotic efficacy of PER NPs in vivo. With IPF progression, injured AEC II gradually died out due to oxidative stress, which leads to mouse suffocation. Hence, restoring normal lung function has important significance. Compared with the BLM group, the PER NPs group had the promising abilities to repair injured lungs and keep them normal. GSH and SOD levels in the PER NPs group were obviously improved with 0.3- and 1-fold, respectively, which could relieve the oxidative stress in injured AEC II to some extent. The level of malondialdehyde (MDA), a key indicator of oxidative stress, was reduced 0.3-fold in the PER NPs group compared with the BLM group (Fig. 4, G to I). Compared with controls, treatment with PER NPs reduced the production of the three cytokines (TGF-, IL-1, and IL-4) in the lungs by 1-, 0.85-, and 0.7-fold, respectively, which showed that the PER NPs effectively inhibited the inflammatory response in IPF lungs (Fig. 4, J to L). We also examined the levels of TGF-, IL-1, and IL-4 in the spleen tissues (fig. S7, B to D), which showed consistent results. These results indicated that PER NPs could treat IPF by inhibiting inflammatory responses in IPF lungs. As seen in Fig. 4M, immunofluorescence staining results revealed that the population levels of fibroblasts CD90+ labeled in green remained in a relatively stable range, while the population levels of activated fibroblasts indicated great proliferation in the BLM group, supporting the conclusion that compared with no treatment group, the PER NPs had an efficient ability to reverse IPF by inhibiting the activation of fibroblasts. Figure 4N showed that the expression of collagen I was notably decreased in the PER NPs group, which confirmed that PER NPs could achieve therapeutic effects by inhibiting ECM deposition.

We next investigated the antifibrosis mechanism based on the synergistic effect of TRA and AST. We firstly established the different formulations, including PLGA-PEG-TRA-AST (PPTA), PLGA-PEG-TRA (PPT), and PLGA-PEG-AST (PPA), and the morphologies of PPA, PPT, and PPTA were evaluated by TEM (fig S8). As shown in Fig. 5A, the results of immunofluorescence staining showed that the expression of the vimentin as cytoskeletal protein was increased after treatment with different formulations in human lung epithelial cell carcinoma (A549). In particular, compared with other treatments, the PER NPs significantly increased the expression of vimentin, indicating that PER NPs had the capacity to keep injured AEC II normal. To assess the repair mechanism induced by the drugs combination, the ROS were detected using ROS probe 2,7-dichlorofluorescin diacetate (DCFH-DA) via inverted fluorescence microscopy and flow cytometry. The ROS content was significant decreased in the PPTA group compared with the untreated and single-drug groups (PPT and PPA) (Fig. 5B). Although the PPA group exhibited some changes than PPT, this effect was not as strong as that in PPTA group, because the PPTA groups exhibited synergistic effect that relieved oxidative stress in injured AEC II than other control formulations. Furthermore, the PPTA group also showed a reduced mitochondrial membrane potential in TGF-induced cells (Fig. 5C), which supported the conclusion that the efficacy in PPTA group was the result of repairing mitochondrial function with relief of oxidative stress in the mitochondria. In the microenvironment of IPF lungs, myofibroblast can be derived from injured AEC II undergoing epithelial-mesenchymal transition (EMT), which aggravates the progression of IPF. As observed in a wound healing assay and invasion assay (Fig. 5D), PPTA effectively inhibited the occurrence of EMT. Furthermore, fibronectin is a structural protein in the ECM, which is a crucial indicator of IPF progression. The expression of fibronectin was obviously decreased in PPTA group than PPT and PPA groups, thus inhibiting the differentiation of injured AEC II into myofibroblast (Fig. 5E). Next, we also tested IPF-reversing efficacy by monitoring the recovery of the lung architecture and improvement in lung functions in vivo. As shown in Fig. 5F, collagen I deposition in the PPTA group returned to normal levels, as determined by H&E and Masson staining, demonstrating that the PPTA could recover the architecture of injured lungs compared with no treatment or single-drug groups (PPT and PPA). Similarly, the expressions of -SMA and collagen I were tested by immunohistochemistry (IHC), which also obtained the same results that the synergistic effect of PPTA could effectively inhibit myofibroblast activation and ECM deposition. In addition, the level of hydroxyproline, the main component of the ECM, was also decreased after treatment with PPTA compared with other treatments (Fig. 5G), suggesting that PPTA had the ability to diminish ECM deposition and retard IPF progression. Furthermore, we detected the expression of -SMA to evaluate the myofibroblasts activation by Western blotting. The lungs were collected after treatment with PPTA, PPT, or PPA for 28 days. The results showed that -SMA expression, as the major evaluation index for IPF, was significantly reduced in PPTA group (Fig. 5H). In addition, the results of real-time quantitative polymerase chain reaction (qPCR) showed that the relative mRNA expressions of CTGF (Ctgf) and -SMA (Acta2) significantly decreased in PPTA group, which indicated that the combination of AST and TRA can achieve efficient therapeutic efficacy by inhibiting myofibroblasts overactivation (Fig. 5, I and J). Moreover, MDA expression decreased, and SOD and GSH levels increased after treatment in PPTA group compared with the PPT and PPA groups. Together, these results implied that the combination of AST and TRA could recover IPF lung function through synergistic effect that was not observed with the other treatments (Fig. 5, K to M). The various formulations as mentioned above were safe by intravenous injection through H&E staining (fig. S9).

(A) Expression of the vimentin in vitro. (B) The ROS level in vitro. (C) The changes of mitochondrial membrane potential. (D) Invasion assay. (E) Fibronectin expression. (F) H&E, Masson, and IHC staining. (G) The level of hydroxyproline. (H) The -SMA and -actin by Western blotting. The mRNA expression of Acta2 (I) and Ctgf (J) by qPCR (n = 3). Contents of GSH (K), MDA (L), and SOD (M) (n = 5). Statistical significance was calculated via one-way ANOVA.

To further investigate the antifibrotic efficacy of MOMC/PER and pirfenidone as a conventional therapeutics for IPF, we evaluated the ability of these treatments to repair lung tissue and inhibit collagen I deposition through H&E and Masson staining, respectively, after 28 days of administration. As shown in Fig. 6A, the alveolar structure in the BLM group collapsed, and alveolar wall thickness increased notably, indicating that collagen I was accumulated and that alveolar heterogeneity was aggravated. Similarly, the alveolar morphologies in the pirfenidone group also showed collapse via H&E staining. In contrast, MOMC/PER could obviously repair the collapsed part of the alveolar space, narrow the spaces between the alveoli, and produce a thinner alveolar wall that tended to appear normal by H&E staining, which demonstrated that MOMC/PER had greater reparative effect on alveolar structure than pirfenidone. In addition, MOMC/PER group showed notable decrease compared with the pirfenidone group in inflammatory cell infiltration. The PER NPs were also more competent in restoring alveolar structure than the clinical drug pirfenidone. This effect was observed because the PER NPs could undergo reprogramming to form MOMC/PER in the blood circulation and then reach their destination, which was consistent with the above results. We further confirmed therapeutic efficacy in regard to ECM deposition by Masson staining (Fig. 6B). Compared with that in the pirfenidone group, the ECM accumulation in the lungs, which appeared as blue staining, was notably reduced in the MOMC/PER group. The results for Masson staining showed that MOMC/PER had greater power than pirfenidone to prevent IPF progression by inhibiting ECM deposition. The main reason for the limited therapeutic effect of pirfenidone was its low bioavailability as an oral drug, and onefold treatment target is the second therapeutic limitation of pirfenidone. Overall, the antifibrotic efficacy in the MOMC/PER group was the best efficacy observed through H&E and Masson staining, and PER NPs had better efficacy than pirfenidone or MOMC. In addition, the fibrosis score of different formulations showed the same trend in fig. S10. These data demonstrated that MOMC/PER showed a preferable combination efficacy over the U.S. Food and Drug Administration (FDA)approved therapeutic pirfenidone or using MOMC or PER NPs alone.

(A) H&E staining. (B) Masson staining. The levels of TGF- in the lungs (C) and spleen tissues (D). N.S., not significant. The levels of IL-1 in the lungs (E) and spleen tissues (F). BUN (G), ALT (H), and aspartate aminotransferase (I) in serum (n = 4). Statistical significance was calculated via one-way ANOVA.

Then, we further evaluated the antifibrotic effect in various groups by examining biochemical indexes of IPF. We first examined the expressions of TGF- and IL-1 in the lungs and spleen tissues, respectively. The results in the lungs showed that TGF- expression was reduced onefold in the MOMC/PER group (P = 0.015) compared with the BLM group and became close to normal (Fig. 6, C and D). However, there was no significant difference between the pirfenidone group and the BLM group, and the level of TGF- in the PER NPs group was lower than that in the pirfenidone group. In addition, the TGF- level in the spleen tissues was significantly decreased in the MOMC/PER group (P = 0.002) compared with the BLM group. However, the level of TGF- in the pirfenidone group was similar to that in the BLM group. The main reason is that pirfenidone is used to treat IPF by inhibiting the accumulation of collagen I, but it has no therapeutic effect on the simultaneous inflammatory response or cytokine expression. The results demonstrated that MOMC/PER had the best antifibrotic efficacy, which was superior to the efficacy achieved by pirfenidone and was mediated by inhibiting the expression of cytokines in the lungs. Furthermore, we detected the expression of IL-1 in the lungs and spleen tissues to investigate the antifibrotic effects of different formulations (Fig. 6, E and F). The trends in IL-1 expression were similar to TGF-; all the treatments could reduce the expression of IL-1, and MOMC/PER showed the best therapeutic efficacy (P = 0.001) in all the treatments. In addition, the IL-1 level in the pirfenidone group was maximal, indicating that compared with the other treatment groups, including the MOMC/PER and PER NPs alone groups, the pirfenidone group showed minimal anti-inflammatory effects. These results indicated that MOMC/PER could achieve a greater treatment effect on IPF than pirfenidone by inhibiting the expression of cytokines in the inflammatory phase; the efficacy of PER NPs was second only to MOMC/PER, and pirfenidone and MOMC were weaker than the PER NPs.

To assess the safety of the treatments in vivo, we then evaluated biological indexes for each formulation after treatment. The levels of blood urea nitrogen (BUN), alanine transaminase (ALT), and aspartate aminotransferase were detected to evaluate the function of the kidneys, liver, and heart, respectively (Fig. 6, G to I). The levels of BUN, ALT, and aspartate aminotransferase were not significantly different between the pirfenidone and other groups (the MOMC/PER, PER NPs, and MOMC groups). As an oral drug approved by the FDA for the treatment of IPF, pirfenidone is highly recognized for its safety in application. Similarly, our different formulations obtained results of safety equivalent to pirfenidone for a certain period of time, indicating that MOMC/PER, MOMC, and PER NPs could also be safely administered by intravenous injection and could be used clinically.

IPF is characterized by injured AEC II and activated myofibroblast, resulting in ECM deposition. To date, the FDA has approved only two drugs (pirfenidone and nintedanib) for IPF treatment. Unfortunately, curing end-stage IPF is inefficient due to the narrow therapeutic spectrums and insufficient accumulation of these drugs in the lungs (3). As a result, traditional therapies have done little to reverse IPF (4). To address this problem, we developed programmed therapeutics MOMC/PER to reverse IPF by efficient lung delivery, programmed modules, and double synergetic strategies.

Two synergetic strategies including drug/drug and cell/drug involved in reversing IPF were shown for the MOMC/PER here. First, the drug/drug as weeding and uprooting strategy could repair injured AEC II and inhibit myofibroblast activation, achieving first synergetic antifibrosis effect. In particular, one drug (AST) acted as the uprooting part of the treatment strategy, repairing injured AEC II by neutralizing oxidative stress. The other drug (TRA) acted as the weeding portion of the strategy, inhibiting the differentiation of fibroblasts into myofibroblast by suppressing CTGF production. Second, some studies have demonstrated that MOMC is multipotent cell that can be specifically recruited to injured lung tissues through interactions between chemokine receptors and chemotactic factors (3638) and contribute to lung tissue normalization and regeneration (39). In addition, MOMC also plays a vital role in regulating the population of immune cells during the inflammatory phase of disease progression (40). Similarly, our results also showed that MOMC could inhibit the proliferation of inflammatory cells, such as lymphocytes and white blood cells. This is another synergetic effect called cell/drug. PER NPs also exhibited greater antifibrotic effects than pirfenidone due to their efficient lung-targeting ability and combination of AST and TRA, as these PER NPs could target MOMC in the circulation, accumulate in the lungs effectively, and then reverse IPF collaboratively. The limited treatment efficacy of pirfenidone is mainly due to its low bioavailability, narrow therapeutic spectrum, and functions by inhibiting myofibroblast activation only.

In addition, MOMC/PER is strategically distinct from nanodelivery carrier (41) and drug-loaded cells carrier (14). The traditional nanodelivery system for IPF always presents dissatisfactory accumulation and unexpected drug release at the lesion site. Even these defects can be avoided for IPF therapy, the therapeutic efficacy is also limited to onefold treating target, and these shortcomings make IPF hard to reverse. In addition to nanodelivery systems, cell-mediated drug delivery has also received more attention in disease treatment. The classic cell-based delivery strategy for treating disease is reliant on drugs being loaded into cells by endocytosis (15). However, cells are difficult to load with large quantities of drugs, and chemotherapeutics may be highly toxic to cells undergoing loading. Hopefully, the PER NPs adhere to the MOMC surface in our study could surmount this challenge in conventional drug loading of cells. PER NPs were firstly attached to MOMC surface and then precisely delivered to the lungs via the homing ability of MOMC and activated for IPF reversion. However, there is still an unresolved point in our research, which is that the treatment mechanism of MOMC remains unclear. Our results indicated that MOMC might up-regulate AEC II proliferation or recover injured AEC II to normalize the lungs. The mechanism of MOMC differentiation for IPF treatment requires further exploration. In addition, some studies have indicated that MOMC could partly treat early IPF through regulating the immune response by inhibiting the proliferation of immune cells in vivo (42). It is unclear whether MOMC is effective for IPF therapy during different periods.

Compared with conventional antifibrotic strategies, our previous unknown programmed therapeutics MOMC/PER has showed accurate lung targeting and excellent therapeutic effects. The excellent antifibrotic efficacy of the MOMC/PER was achieved through the following features. (i) MOMC has the ability to backpack PER NPs, constructing programmed therapeutics MOMC/PER. (ii) MOMC/PER can precisely accumulate in IPF lung tissues due to the homing ability of the MOMC. (iii) PER NPs are sensitively released from MOMC/PER due to the overexpression of MMP-2 in the IPF microenvironment. (iv) Released PER NPs are able to retarget injured AEC II through c(RGDfc). (v) PER NPs can reduce the secretion of TGF- by occupying TGF-latent sites. (vi) Two drugs loaded into PER NPs are the key factors in achieving IPF reversion of drug/drug as weeding and uprooting. In addition, MOMC also participates in AEC II regeneration using cell/drug strategy. Specifically, MOMC-mediated delivery therapeutics is convenient, and the materials used in our PER NPs have all been approved by the FDA, which indicates certain advantages for further clinical development. Overall, we have proposed an innovative concept to cure IPF through using native cells as a delivery carrier and a dual-drug combination as therapeutic agents, and this strategy is likely to be applicable to other major diseases.

MMP-2, -SMA rabbit anti-mouse antibody, and DCFH-DA were purchased from Sigma-Aldrich (St. Louis, USA). SPC rabbit anti-mouse antibody was purchased from Millipore (St. Louis, USA). Lymphocyte isolation kit was purchased from Solarbio Science & Technology Co. Ltd. (Beijing, China). PLGA-PEG-Mal and PLGA-PEG-c(RGDfc) were purchased from Jinan Daigan Biomaterial Co. Ltd. (Jinan, China). RPMI 1640, fetal bovine serum (FBS), and bicinchoninic acid (BCA) protein assay kit were purchased from Jiangsu KeyGEN BioTECH Co. Ltd. (Nanjing, China). The peptide E5 (CGPLGIAGQCGGRSFFLLRRIQGCRFRNTVDD) was synthesized by Top Peptide Biotechnology Co. Ltd. (Shanghai, China). AST was purchased from Yuanye Bio-Technology Co. Ltd. (Shanghai, China). TRA was purchased from J&K Scientific Co. Ltd. (Beijing, China). DiI, 3,3-dioctadecyloxacarbocyanine perchlorate (DiO), and DiR were purchased from Fanbo Biochemicals Co. Ltd. (Beijing, China). DAPI (4,6-diamino-2-phenylindole) and mitochondrial membrane potential kit of JC-1 were purchased from Beyotime Biotechnology Co. Ltd. (Shanghai, China). CXC chemokine ligand 12 (CXCL 12) and CC chemokine ligand 19 (CCL 19) were purchased from Zoonbio Biotechnology Co. Ltd. (Beijing, China). BLM was purchased from Zhejiang Huahai Pharmaceutical Co. Ltd. (Linhai, China). TGF- was purchased from Multi Sciences Biotech Co. Ltd. (Hangzhou, China). C6 was purchased from Tokyo Chemical Industry Co. Ltd. (Tokyo, Japan). LysoTracker Red DND-99 kit was purchased from Thermo Fisher Scientific (Waltham, USA). Nanog, TGF-/Smad and collagen I rabbit anti-mouse antibodies, and H&E and Masson staining kits were purchased from Servicebio Co. Ltd. (Nanjing, China). GSH, MDA, SOD, and hydroxyproline were purchased from Jiancheng Biotech Co. Ltd. (Nanjing, China). IL-1 and IL-4 detection kits were purchased from eBioscience (Waltham, USA). TGF- detection kit was purchased from BioLegend (CA, USA). Polyvinylidene difluoride (PVDF) was purchased from PALL (NY, USA). Electrogenerated chemiluminescence (ECL) was purchased from Tanon Science & Technology Co. Ltd. (Shanghai, China).

MOMC was isolated from peripheral blood by a mouse lymphocyte isolation kit. To obtain the MOMC, peripheral blood was collected from the C57BL/6J mice of IPF with EDTA and diluted three times with phosphate-buffered saline (PBS). The isolated protocol of MOMC was as follows: The mouse Percoll was added into diluted peripheral blood solution, and MOMC was isolated from peripheral blood by centrifugation at 600 rpm for 30 min. The solution was divided into upper, middle, and lower layers, and MOMC existed in the middle layer. Then, the MOMC was taken into centrifuge tube of 15 ml, and cell washing buffer of 5 ml was added into the tube. The cell suspension was then centrifuged for another 30 min, and it needed to be repeated three times to obtain MOMC. Last, the cell deposits were resuspended, and the suspension was put into the culture dish in RPMI 1640 medium with 10% FBS at 37C and 5% CO2 (22, 23). The adherence time for dishes of MOMC was about 2 weeks. The MOMC was a kind of adherent cells, and the morphologies of cells were fusiform. After the cells adhere to the dish, the medium was changed once every 3 days.

PER NPs were prepared by antisolvent precipitation method. Peptide E5 modification was prepared as follows. Specifically, the PLGA-PEG-Mal and PLGA-PEG-c(RGDfc) (mass ratio, 10:1) and dual drugs of AST and/or TRA were dissolved in dimethyl sulfoxide (50 mg/ml, 2 ml), added dropwise into deionized water with 100 ml, and then stirred with 300 rpm for 2 hours. Next, the prepared nanoparticle solution (NPs) was centrifuged at 2800 rpm for 15 min to discard the large particles and free drug. The NPs were then condensed to concentration of 2 ml by ultrafiltration device for further use. Last, the peptide E5 was added into the solution of NPs to form PER NPs. The preparation process of other groups including PLGA-PEG-c(RGDfc) (PR NPs), the preparation of PLGA-PEG-E5 (PE NPs), and the preparation of PLGA-PEG (PP NPs) were similar to PER NPs.

The preparation of MOMC/PER was carried out by incubating MOMC with PER NPs. Briefly, the MOMC (2 105 cells/ml) was cultured in a petri dishes with a diameter of 100 mm. After incubated with the FBS-free media for 1 hour, PER NPs at a TRA concentration of 40 g/ml were added into MOMC medium and incubated for 2 hours at 37C and 5% CO2. At the same time, the CXCR4 receptor and ligand peptide E5 would undergo bioconjugate reaction.

The hydrodynamic diameters and potentials for PER NPs suspended in 1 PBS were measured by Brookhaven Instruments (NY, USA). The morphologies of PER NPs were characterized by TEM (Hitachi TEM system, Japan).

For MOMC/PER-DiI, characterization of adhesion between MOMC and PER-DiI NPs was imaged by CLSM (Carl Zeiss 700, Germany). Specifically, MOMC was cultured in 35-mm culture dishes and incubated with PER-DiI NPs for 2 hours, and the preparation of PER-DiI NPs was the same as mentioned above. The nucleus of MOMC was labeled with DAPI and MOMC membrane was labeled by DiO. For SEM characterization, MOMC/PER-DiI was coated with gold/palladium and examined by Hitachi-SU8020 (Japan).

The sensitive release properties of PER NPs from MOMC/PER in vitro was evaluated under the microenvironment of MMP-2 in vitro. MMP-2 enzyme was applied to release PER NPs by degrading the linker of GPLGIAGQ between PER NPs and MOMC. The characterization of released activity was investigated in vitro. First, MMP-2 (2 g/ml, 1 ml) was added into MOMC/PER-DiI medium for 30 min. Then, MOMC and released PER-DiI NPs were detected by flow cytometry (BD FACSCalibur, USA) or fixed by 4% paraformaldehyde (w/v) for CLSM and 2.5% glutaral for TEM. Besides, the nucleus of MOMC was labeled with DAPI for 30 min at 37C, and the MOMC membrane was labeled with DiO for 15 min at 37C. MMP-2 enzyme was dissolved in deionized water and free RPMI 1640 (volume ratio, 1:20) at a concentration of 2 g/ml. After that, the released PER NPs and MOMC in solution were prepared for the other testing assay and image.

The loading ability of MOMC was detected by the concentration of TRA and AST. First, MOMC was cultured in dishes with 100 mm. After MOMC adhered on the dishes, PER NPs were added into the culture dishes and incubated with MOMC for 2 hours in RPMI 1640 with free FBS, and then, MOMC/PER was washed thrice with PBS and digested with 0.25% trypsin-EDTA solution. Next, cells were harvested by centrifugation at 1000 rpm for 3 min and counted with a hemocytometer. Last, the cells were resuspended with PBS of 1 ml, and the absorbance was determined at 326 nm for TRA and 491 nm for AST by Multiskan GO (Thermo Fisher Scientific, USA).

The migration capacity of MOMC/PER was investigated by a Transwell device. First, the MOMC were cultured into the upper chambers with pore sizes of 8.0 m with 2 104 in 400 l of RPMI 1640 with FBS-free media for 24 hours. The cells were divided into three groups, including CXCL 12 () or CCL 19 () of MOMC, CXCL 12 (+) or CCL 19 (+) of MOMC, and CXCL 12 (+) or CCL 19 (+) of MOMC/PER. RPMI 1640 with 10% FBS and CXCL 12 (10 g/ml) or CCL 19 of 600 l was added to the lower chamber into the 24-well plates. Then, the MOMC in MOMC/PER group was added PER NPs at concentration of 40 g/ml and incubated for another 24 hours. At last, the Transwell chambers were stained with crystal violet and were dissolved with 33% acetic acid, and the absorbance of solution was tested at 570 nm.

C57BL/6J male mice were obtained from East China Normal University Laboratory Animal Technology Co. Ltd. (Shanghai, China) and housed with a 12-hour light/12-hour dark cycle at 25C. All the animal protocols and procedures were performed under the guidelines for human and responsible use of animals in research approved by the regional ethics committee of China Pharmaceutical University. After acclimatization for 7 days, mice were subjected to IPF model experiments. IPF mice models were established by inhalation of BLM through endotracheal intubation (2 U/kg, 40 l; Braintree Scientific, USA). Next, the mice were randomly assigned to the treatment. For the cell culture, A549 and MOMC were cultured in RPMI 1640 media containing 10% FBS and 1% penicillin and streptomycin at 37C and 5% CO2.

A549 cells were cultured in six-well plates at 37C and 5% CO2 for 24 hours and then incubated for another 24 hours with pure PPA, PPT, or PPTA at TRA concentration of 20 nM. The expression of vimentin and fibronectin was investigated by immunofluorescence staining.

The invasion ability of TGF-induced A549 cells was evaluated by wound healing tests after treated with various formulations. First, A549 cells were seeded in six-well plates at 15 104 cells per dish and incubated for 24 hours. Next, a 10-l pipette tip was used to scratch wells in the middle of the dishes, and then, A549 cells were washed three times with PBS to remove suspended cells. The cells from each group were imaged by inverted fluorescence microscope (Nikon, Japan) to observe the extents of wound healing after treated with PPT, PPA, PPTA, and PER NPs at 24 hours.

The migration capability of TGF-induced A549 cells was investigated by a Transwell device. The A549 cells of 5 104 in 400 l of RPMI 1640 with FBS-free media were added to the upper chambers with pore sizes of 8.0 m for 24 hours, and RPMI 1640 with 10% FBS media of 600 l was added to the lower chamber into the 24-well plates. Then, the cells were incubated with PPT, PPA, PPTA, and PER NPs (20 nM of TRA concentration) for another 24 hours. After incubation, the Transwell chambers were stained with crystal violet and were dissolved with 33% acetic acid, and the absorbance of solution was tested at 570 nm.

A549 cells were seeded on six-well plates (15 104 per well) and incubated overnight. First, the cells were activated by TGF-, and then, various treatment groups were incubated with A549 cells for 24 hours (TGF-, normal, PPT, PPA, PPTA, and PER NPs). Next, the ROS probe DCFH-DA (5 M) was added into the dishes and incubated with cells in RPMI 1640 media with free FBS at 37C under 5% CO2 in the dark for 15 min. Last, cells were digested, and the content of ROS was analyzed by flow cytometry (BD Accuri C6, USA) and imaged by inverted fluorescence microscope, respectively.

A549 cells were cultured on six-well plates (15 104 per well) overnight at 37C and 5% CO2. After treated with different formulations for 24 hours, 500 l of mitochondrial membrane potential reagent of JC-1 (1) solution was added into the dishes for 20 min. Then, the cells were stained by 4% paraformaldehyde (w/v) and imaged by inverted fluorescence microscope.

First, A549 cells were seeded on a 35-mm sterile glass bottom culture dishes (2 105 cells) and cultured overnight in RPMI 1640 with 10% FBS. The preparation of PR-C6 and PP-C6 was same as mentioned above for PER NPs. The PR-C6, PP-C6, and C6 were then incubated with A549 cells for 4 hours at 5 g/ml. Next, the cells were washed three times by PBS, and the nucleus was stained with DAPI. Images and data were acquired with CLSM and flow cytometry (BD Accuri C6, USA).

The A549 cells of 5 104 were cultured in a 35-mm sterile glass bottom culture dishes. After the cells were cultured overnight in RPMI 1640 with 10% FBS for 24 hours, PR-C6 and PP-C6 were incubated with A549 cells for 1 and 4 hours at 5 g/ml. Then, the cell dishes were washed three times by PBS and fix by 4% paraformaldehyde (w/v). Lysosome was stained with LysoTracker Red DND-99 kit (100 nM) for 15 min, and the dishes were washed three times with PBS. Then, the cell nucleus was stained by DAPI like above method. Images were acquired by CLSM.

MOMC/PER-DiR was prepared by the method as mentioned above for PER NPs. The homing ability of MOMC was detected in vivo by IVIS imaging system (Kodak, USA). C57BL/6J mice were induced IPF by inhalation of BLM. Then, the IPF mice were injected with MOMC/PER-DiR, MOMC-DiR, and free DiR by intravenous injection and tested by IVIS living system at different point times. Then, the mice are sacrificed, and the lungs and other organs were harvested for ex vivo imaging after 8 hours of intravenous injection. ROI was circled around the lungs and the other organs (liver, heart, spleen, and kidneys). The fluorescence intensity of the DiR was determined by living image software.

The PER NPs can be released from MOMC/PER-DiI by MMP-2, owing to responsive blocking the linker between PER NPs and MOMC. We have investigated the homing capability, responsive release, and retarget ability. We first administrated MOMC/PER-DiI by intravenous injection, and the preparation of MOMC/PER-DiI was applied by the methods as mentioned above. The mice were sacrificed, and lung tissues were harvested at different point time of 30 min, 1 hour, and 2 hours in a dark place. First, the lung tissues were fixed with 4% paraformaldehyde (w/v), and then, the tissues were embedded and dewaxed before slicing. Next, the lung slices were labeled by Nanog and SPC at 4C for 1 hour and washed with 0.2% Triton X-100 for three times. Then, they were incubated with relevant secondary antibodies for 2 hours. Thereafter, the slices were stained with DAPI and viewed under fluorescence microscope.

The IPF mice were injected with PER-DiR NPs, PE-DiR NPs, PR-DiR NPs, and PP-DiR NPs by intravenous injection at different point times for 1, 4, 8, 12, and 24 hours and tested by IVIS imaging system. The preparation of PER-DiR NPs, PE-DiR NPs, PR-DiR NPs, and PP-DiR NPs were the same method as mentioned above, and then, the mice were sacrificed, and the lungs and other organs were harvested to detect ex vivo imaging after intravenous injection at 24 hours. ROI was tested on the lungs and the other organs (liver, heart spleen, and kidneys). The fluorescence intensity of the DiR was determined by living image software.

The antifibrotic efficacy in MOMC/PER, MOMC, PER NPs, and pirfenidone for IPF treatment in vivo was evaluated on IPF male mice (C57BL/6J, age of 6 to 8 weeks). The preparations of MOMC/PER and PER NPs were the same method as mentioned above. Pirfenidone was administered by gastrointestinal because it is an oral medication, and other formulations were administered via intravenous injection.

The contents of MDA, SOD, and GSH were detected after treated with various treatments in lung tissues. The protocols are as follows: Solution 1 of 1.5 ml was added into the lung tissues solution (0.5 ml) and mixed thoroughly. Then, the samples were centrifuged for 10 min at 3500 to 4000 rpm. The sample supernatant was added into 3,3,5,5-tetramethyl benaidine (TMB substrate), and the absorbance of GSH was detected after 5 to 10 min at 420 nm. The contents of SOD and MDA were tested at 550 and 532 nm, respectively.

The lungs and spleen tissues were collected and diluted in precooled solution. The IL-1, TGF-, and IL-4 in lung tissues were assayed using enzyme-linked immunosorbent assay (ELISA) method as instructed by the manufacturer. First, the wells were washed three times with a washing buffer for 3 min each time. Next, the blocking solution of 200 l was added into each well and incubated for 1 to 2 hours at 37C. Then, the sealing film was removed carefully and putted it into the washing machine and washed three to five times. Furthermore, the sample of 100 l was added and should be tested diluted appropriately to the above coated reaction wells, and the diluted biotinylated antibody working solution was added with 100 l into each well. Then, the samples were sealed with a sealing membrane and incubated at 37C for 1 hour. The following step was that 100 l of diluted enzyme conjugate working solution was added into each well. Next, TMB substrate solution with 100 l was added into each well and should be avoided reaction with light for 10 to 30 min at 37C until a notable color gradient appears in the diluted standard well. Within 10 min, the absorbance of each well was measured on a microplate reader at 450 nm with zero adjustment of the blank control well.

The content of hydroxyproline was detected by a hydroxyproline detection kit. The lung tissues of 30 to 100 mg were mixed with hydrolysate to 1 ml and hydrolyzed in boiling water for 20 min. The sample solution was pH 6.0 to 6.8. Then, the serum hydrolysate was added activated carbon and mixed at 60C for 15 min. After cooling, the serum samples were centrifuged at 3000 rpm for 20 min, and the supernatant was detected by microplate reader at 550 nm.

The whole blood from mice was collected by anticoagulant tube with EDTA, and white blood cell counts (including lymphocytes and monocytes) were assayed using a standard blood analyzer (Mindray, China).

After treatment with different formulation, the lungs, heart, liver, spleen, and kidneys were harvested, and lungs were investigated by H&E, Masson, and IHC staining. Other organs were detected by H&E staining to evaluate the application security. First, the lung tissues were fixed with 4% paraformaldehyde (w/v) for more than 48 hours and embedded in paraffin. Then, the lung tissues were cut into 4-m sections for H&E staining and Masson staining. The levels of collagen I and -SMA were evaluated by IHC, and protocols are as follows: The slices were incubated with primary antibodies (collagen I and -SMA or fluorescently labeled CD90 and collagen I, Servicebio, China) and then incubated with corresponding secondary antibodies to detect the expression of relative proteins.

The qPCR analysis was evaluated RNA expression of Ctgf and Acta2. qPCR was conducted in ABI StepOnePlus (Thermo Fisher Scientific, USA). Lung tissues (100 mg) were homogenized to extract the total RNA according to the protocols. Complementary DNA (2 g) was prepared using the Reverse Transcription System, and then, the expression of related genes was determined using q-PCR. The primers used are Acta2 (NM_007392.3), Ctgf (43), and Gapdh (NM_008084.2).

The harvested lungs were homogenized in PBS buffer and then centrifuged at 3000 rpm for 30 min. The supernatant of total protein was taken for further experiments. Total protein concentration in the solution was determined with a BCA protein assay kit. After detecting in SDS-PAGE with protein samples in different treatment groups, the bands were transferred onto PVDF membrane. Next, the PVDF membranes were blocked with 5% milk at room temperature for 2 hours and incubated with primary antibodies (-SMA, TGF-/Smad, and -actin rabbit anti-mouse antibodies) at 4C overnight and then incubated with corresponding secondary antibodies for 2 hours at room temperature. Last, the bands were detected using ECL (Tanon, China) Western blotting substrate (Thermo Fisher Scientific, USA). The -actin was used as an endogenous control.

The mice were sacrificed, and the serum samples were detected in different treatment groups. The contents of ALT, aspartate aminotransferase, and BUN in the serum were determined using the relevant assay kits (Servicebio, China).

Statistical analyses were performed using GraphPad Prism software (GraphPad Software, USA). All error bars were means SEM; differences detection index between the treated groups and control groups were determined via one-way analysis of variance (ANOVA). P < 0.05 was considered significantly different.

Acknowledgments: We thank the Cellular and Molecular Biology Center of China Pharmaceutical University for assistance with confocal microscopy work. Funding: This work was supported by the National Key R&D Program of China (2017YFA0205400). We thank the National Natural Science Foundation of China (NSFC; grant nos. 81773667, 81573369, and 81430082) and NSFC Projects of International Cooperation and Exchanges (81811540416). This work was also supported by the Fundamental Research Funds for the Central Universities (2632018PT01 and 2632018ZD12), the 111 Project from the Ministry of Education of China and the State Administration of Foreign Experts Affairs of China (B16046), the Double First-Class Project (CPU2018GY06), and a project funded by the Priority Academic Program Development of Jiangsu Higher Education Institutions. Author contributions: H.-L.J., H.-P.H., X.C., and L.X. conceived the project, designed all the experiments, analyzed the data, and wrote the manuscript. X.C. and L.X. conducted the experiments. X.C. and Y.W. analyzed the data. X.C., Y.W., C.-X.Y., Y.-J.H., T.-J.Z., X.-D.G., and L.L. wrote the manuscript. All authors edited the manuscript. 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 this article:
Monocyte-derived multipotent cell delivered programmed therapeutics to reverse idiopathic pulmonary fibrosis - Science Advances