Small Leucine-Rich Proteoglycans (SLRPs) Are Involved in the Anti-Myeloma Response of Osteoblasts.

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 815-815
Author(s):  
Xin Li ◽  
Angela Pennisi ◽  
Shmuel Yaccoby

Abstract Multiple myeloma (MM) is closely associated with severe osteolytic bone disease caused by stimulation of osteoclastogenesis and suppression of osteoblastogenesis. We have recently demonstrated that while osteoclasts support MM growth, osteoblasts have negative impact on survival and proliferation of MM cells from a subset of patients in vitro and in vivo (Yaccoby et al., Cancer Res 2004: Haematologica 2006), indicating that MM bone disease drives tumor progression. To unravel anti-MM molecular mechanism of osteoblasts we performed global gene expression profiling on osteoblasts and their precursors, mesenchymal stem cells (MSCs). We found and validated by qRT-PCR and Western Blotting that osteoblasts produce high level of small leucine-rich proteoglycans (SLRPs). Main SLRPs such as decorin, biglycan and lumican are essential for proper bone remodeling have been shown to inhibit growth of various tumors and their expression is inversely correlated with tumor progression. Decorin is not produced by most MM cases and recombinant decorin was rapidly internalized by MM cells. Co-culture of MM cells with the supporting osteoclasts in the presence of osteoblast’s conditioned media resulted in reduced MM cell survival (n=5, p<0.002), an effect that was partially but significantly attenuated by decorin neutralizing antibody (5 μg/ml, n=5, p<0.01). In co-culture with osteoblasts, primary MM cell growth was significantly increased by 46±15% when decorin expression in osteoblasts was effectively blocked using lentiviral-containing decorin siRNA (n=4, p<0.04). In contrast overexpression of decorin in MSCs resulted in reduced primary MM cell survival by 20±3% (n=4, p<0.04). Furthermore, in co-culture with osteoclasts recombinant or purified decorin (5–10 μg/ml) inhibited growth of primary MM cells in 6 of 9 experiments by 35±8% (p<0.007). The anti-MM effect of decorin involved direct induction of apoptosis as determined by annexin V/PI flow cytometry and activation of p21 using Western Blotting. In additional experiments we demonstrated that conditioned media from culture of MM cells stimulated tube formation by HUVECs in a matrigel assay and that decorin significantly attenuated this effect (2±1 vs. 12±1 tubes in control wells, p<0.001), indicating that certain SLRPs are antiangiogenic. Decorin (5–10 μg/ml) also effectively inhibited differentiation of osteoclasts in culture of osteoclast precursors with RANKL and M-CSF (14±2 vs. 32±4 multinucleated TRAP-expressing osteoclasts in control wells, p<0.01). For in vivo study we exploited our SCID-hu model for primary MM (Yaccoby et al., Blood 1998: 1999). Immunohistochemical analysis for decorin and lumican in nonmyelomatous human bone implanted in SCID-hu mice revealed high expression level by osteogenic cells and localization along the bone surface and extracellular matrix. Decorin and lumican levels were reduced in myelomatous bones engrafted with primary MM cells and markedly increased following treatment with osteoblast activating agents such as PTH and anti-DKK1, providing a possible molecular mechanism for anti-MM efficacy of these agents in vivo (Yaccoby et al., Blood 2007). We conclude that certain SLRPs are involved in the anti-MM effect of mature osteoblasts directly and indirectly through inhibition of angiogenesis and osteoclastogenesis, and that increasing endogenous or exogenous SLRPs in myelomatous bones are safe approaches to control MM and its associated bone disease.

Author(s):  
Beatriz Gamez

Gámez B., Morris EV., Olechnowicz S., Sowman, A., Turner, C. and Edwards CM.   Multiple myeloma (MM) is a fatal malignancy characterized by an expansion of malignant plasma cells in the bone marrow (BM) and associated with osteolytic bone disease. MM is preceded by the benign condition, monoclonal gammopathy of undetermined significance (MGUS). Understanding MGUS progression and development of MM bone disease is key for patient management. We and others have previously demonstrated that diet-induced obesity promotes myeloma progression, but the mechanisms underlying this remain unknown. The aim of the current study was to determine the effect of dietary cholesterol on MM development. A 2% cholesterol diet was used to increase circulating LDL in mice. Mice were randomly distributed to either a) cholesterol diet 4 weeks prior to 5TGM1 MM inoculation (pretreatment) or b) cholesterol diet 4 weeks prior to MM inoculation and continued for the entire experiment (continuous). Mice on the continuous cholesterol diet had increased tumour burden, associated with an increase in lipid droplet content of MM cells. No differences in tumour burden were seen in those mice where cholesterol diet was halted at time of MM inoculation. In vitro, myeloma cells cultured with delipidated FBS had a 50% reduction in viability after 72 hours. Rich cholesterol content lipoproteins (LDL) but not VLDL could restore MM cell viability, suggesting that cholesterol is responsible for this lipid-depletion effect. Taken together, our results show that high cholesterol promotes myeloma and results in a higher lipid content in myeloma cells, ultimately increasing BM tumour burden. Pretreatment with a cholesterol diet did not alter disease progression suggesting a direct pro-tumourigenic effect of cholesterol. These results demonstrate both the detrimental effect of cholesterol on myeloma progression and the potential for dietary intervention approaches.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 2503-2503
Author(s):  
Sonia Vallet ◽  
Noopur Raje ◽  
MariaTeresa Fulciniti ◽  
Kenji Ishitsuka ◽  
Teru Hideshima ◽  
...  

Abstract Osteolytic bone disease (OBD) is a frequent complication of multiple myeloma (MM), affecting 70 to 80% of the patients. OBD is characterized by imbalanced bone remodeling, due to decreased osteoblast (OB) number and increased osteoclast (OC) formation and activity. MM cells secrete osteoclastogenic factors, such as receptor activator of nuclear factor kappa B ligand (RANKL) and CCL3. In turn, OC support MM cell proliferation and survival, thus promoting a positive feedback that exacerbates bone resorption. Chemokines modulate osteoclastogenesis and promote MM cell proliferation, in particular CCL3 and its receptor CCR1 play an important role in mediating OBD in MM. MLN3897 (Millennium Pharmaceuticals, Cambridge) is a novel small molecule specific antagonist of human CCR1 (IC50 0.8 nM). It has a favorable toxicity profile in healthy volunteers and is currently undergoing phase II clinical trials in rheumatoid arthritis and multiple sclerosis. Here we evaluate the effects of MLN3897 on OC function and activity, as well as OC-MM cell interactions. Our in vitro data demonstrates a dual mechanism of action for MLN3897: it inhibits osteoclastogenesis and also overcomes the protective effects conferred by OC on MM cells. Our data further shows inhibition of OC formation and function by 40 and 70%, respectively, following MLN3897 treatment. This is mediated via inhibition of the fusion process and is accompanied by downregulation of pERK and c-fos signaling. To analyze its effect on MM cells, we verified CCR1 and CCR5 expression levels on MM1.S (15% and 3.6%) and OPM1 (3.8 and 0.7%). Our data show that OC secrete high levels of CCL3 which triggers MM cell migration; and that MLN3897 abrogates these effects by inhibiting the PI3K/Akt pathway. Moreover, MLN3897 overcomes the proliferative advantage conferred by OC on MM cells, as demonstrated in INA6, MM1.S and MM patient derived primary cells. OC induced MM cell proliferation is mediated by adhesion and cytokine secretion, and MLN3897 abrogates both MM cell-to-OC adhesion and interleukin-6 (IL6) secretion by OC in a co-culture system, thereby resulting in decreased MM cell survival and proliferation. To confirm these in vitro results, in vivo studies in a SCID-hu mouse model are underway. Implanted SCID-Hu INA-6 bearing mice are treated with twice daily oral MLN3897 for 3 weeks. The evaluation of osteolytic lesions and OC, OB and endothelial cell number; and tumor burden will be presented. Our in vitro results therefore show novel biologic sequelae of CCL3 and its inhibition on both osteoclastogenesis and MM cell growth. Our in vivo experiments will further validate the role of CCR1 in a human BM microenvironment-MM model, providing the framework for clinical trials of MLN3897 for the treatment of OBD in MM.


Author(s):  
Beatriz Gamez

Gámez B., Morris EV., Olechnowicz S., Sowman, A., Turner, C. and Edwards CM. Multiple myeloma (MM) is a fatal malignancy characterized by an expansion of malignantplasma cells in the bone marrow (BM) and associated with osteolytic bone disease. MM ispreceded by the benign condition, monoclonal gammopathy of undetermined significance(MGUS). Understanding MGUS progression and development of MM bone disease is key forpatient management. We and others have previously demonstrated that diet-induced obesitypromotes myeloma progression, but the mechanisms underlying this remain unknown. The aimof the current study was to determine the effect of dietary cholesterol on MM development. A2% cholesterol diet was used to increase circulating LDL in mice. Mice were randomlydistributed to either a) cholesterol diet 4 weeks prior to 5TGM1 MM inoculation (pretreatment)or b) cholesterol diet 4 weeks prior to MM inoculation and continued for the entire experiment(continuous). Mice on the continuous cholesterol diet had increased tumour burden, associatedwith an increase in lipid droplet content of MM cells. No differences in tumour burden wereseen in those mice where cholesterol diet was halted at time of MM inoculation. In vitro,myeloma cells cultured with delipidated FBS had a 50% reduction in viability after 72 hours. Richcholesterol content lipoproteins (LDL) but not VLDL could restore MM cell viability, suggestingthat cholesterol is responsible for this lipid-depletion effect. Taken together, our results showthat high cholesterol promotes myeloma and results in a higher lipid content in myeloma cells,ultimately increasing BM tumour burden. Pretreatment with a cholesterol diet did not alterdisease progression suggesting a direct pro-tumourigenic effect of cholesterol. These resultsdemonstrate both the detrimental effect of cholesterol on myeloma progression and thepotential for dietary intervention approaches.  


Cancers ◽  
2021 ◽  
Vol 13 (24) ◽  
pp. 6327
Author(s):  
Adi Knigin ◽  
Shani Avniel-Polak ◽  
Gil Leibowitz ◽  
Kira Oleinikov ◽  
David J. Gross ◽  
...  

(1) Background: Neuroendocrine neoplasms of the lung (LNENs, lung carcinoids) are often diagnosed at an advanced stage when they are not surgically curable, and treatment options are limited. One of the approved options for treating inoperable tumors is everolimus—an mTOR inhibitor (mTORi). Activation of mTOR, among many other effects, inhibits autophagy, which is a cell survival mechanism in general, and in tumor cells in particular. Everolimus may paradoxically encourage cancer cell survival. In practice, the drug inhibits tumor development. Chloroquine (CQ) is a known antimalarial compound that inhibits autophagy. Our research is focused on the hypothesis that autophagy plays a key role in the development of tumor resistance to mTORi, and that the addition of autophagy inhibitors to mTORi exerts a synergistic effect on suppressing tumor cell proliferation. We have recently demonstrated that the combination of CQ with different mTORi increases their potency compared with mTORi alone in both in vitro and in vivo models of pancreatic NENs. In this study, we examined the effects of CQ and mTORi on in vitro and in vivo LNEN models. Aims: Testing the effects of CQ together with mTORi on cell proliferation, apoptosis, and autophagy in in vitro and in vivo LNEN models. (2) Methods: The NCI-H727 LNEN cells were treated with CQ ± mTORi. Cells’ viability and proliferation were measured using XTT and Ki-67 FACS staining. The effects of the treatments on the mTOR pathway and autophagy were examined using Western blotting. Cytotoxicity was measured using a cytotoxicity kit; apoptosis was measured by PI FACS staining and Western blotting. We further established an LNEN subcutaneous murine xenograft model and evaluated the effects of the drugs on tumor growth. (3) Results: CQ alone suppressed LNEN cells’ viability and proliferation and increased their cytotoxicity and apoptosis; these effects were augmented when CQ was added to an mTORi. We also showed the possible mechanisms for these results: on the one hand we could see a decrease in P62 levels and the absence of LC3-II (both inversely related to autophagy) following treatment with the mTORi, and on the other hand we could demonstrate an increase in their levels when CQ was added. The effect was less apparent in the murine xenograft model. (4) Conclusions: By inhibiting autophagy and inducing apoptosis, CQ suppresses tumor cell growth in LNENs. CQ potentiates mTORi effects, implying that further studies are needed in order to elucidate its possible role in tumor inhibition in patients with LNENs.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 812-812
Author(s):  
Claire M. Edwards ◽  
James R. Edwards ◽  
Seint T. Lwin ◽  
Gregory R. Mundy

Abstract Multiple myeloma is characterized by uncontrolled proliferation of myeloma cells within the bone marrow and the development of a severe osteolytic bone disease. In addition to a well characterized increase in osteoclastic bone resorption, myeloma bone disease is associated with a reduction in bone formation. Osteoblast differentiation and bone formation are regulated in vivo by canonical Wnt signaling and activation of β-catenin. Therefore increasing Wnt signaling in the bone microenvironment in multiple myeloma may prevent the development of myeloma bone disease. In support of this, we have previously demonstrated that activation of Wnt signaling with lithium chloride (LiCl) in the 5TGM1 murine model of myeloma reduces tumor burden and osteolytic bone disease. However, we also found that LiCl treatment increased subcutaneous (s.c.) tumor growth. This suggests that the reduction in tumor burden within the bone microenvironment may be an indirect effect mediated through the effects of LiCl to prevent myeloma bone disease. The aim of the current study was to determine the effect of specific molecular blockade of Wnt signaling in myeloma cells in vivo. 5TGM1-GFP myeloma cells were transfected by electroporation with either myc-tagged dominant negative TCF4 (DNTCF4) or pcDNA. Following stable selection by culture in G418, expression of DNTCF4 was confirmed by western blot for myc. No difference was found in the growth rates of 5TGM1-pcDNA or 5TGM1-DNTCF4 in vitro. Treatment with LiCl or Wnt3A had no significant effect on cell viability in vitro, but significantly increased β-catenin activity, as measured by TOPFLASH activity in 5TGM1-pcDNA cells. This increase was not observed in 5TGM1-DNTCF4, confirming that expression of DNTCF4 blocked Wnt signaling induced by LiCl in 5TGM1 myeloma cells. C57Bl/KaLwRij mice were inoculated with 5TGM1-pcDNA or 5TGM1-DNTCF4 cells by either intravenous (i.v.) or s.c. injection. Mice were treated from time of tumor cell inoculation with 200mg/kg/day LiCl or vehicle control (d.H20) by oral gavage for 28 days. I.v. inoculation of myeloma cells resulted in a significant increase in serum IgG2bκ concentrations and the proportion of GFP-positive cells in the bone marrow. A significant reduction in trabecular bone volume was also observed. MicroCT analysis of the tibia demonstrated that LiCl significantly increased trabecular bone volume in both 5TGM1-pcDNA and 5TGM1-DNTCF4 myeloma-bearing mice. LiCl significantly decreased serum IgG2bκ concentrations in both 5TGM1-pcDNA and 5TGM1-DNTCF4 myeloma-bearing mice, with a greater effect in 5TGM1-DNTCF4 myeloma-bearing mice. FACS analysis of GFP-positive cells demonstrated that LiCl significantly reduced tumor burden in the bone marrow in both 5TGM1-pcDNA and 5TGM1-DNTCF4 myeloma-bearing mice. However, following s.c inoculation, LiCl significantly increased s.c. tumor volume of 5TGM1-pcDNA tumors, but had no effect on 5TGM1-DNTCF4 s.c. tumor volume. Taken together these results demonstrate that the effect of increasing Wnt signaling in myeloma is dependent upon the microenvironment. By specific inhibition of β-catenin activity in myeloma cells combined with systemic stimulation of the Wnt signaling pathway, our results suggest that increasing Wnt signaling in myeloma in vivo has dual effects; firstly to enhance myeloma growth directly, and secondly to enhance osteoblast differentiation and thus indirectly reduce tumor burden in bone, highlighting the importance of the bone marrow microenvironment in regulating myeloma growth and survival.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 3154-3154
Author(s):  
Davorka Messmer ◽  
Hsu-Hsiang Chang ◽  
Thomas J. Kipps

Abstract The tumor microenvironment plays a critical role in growth, metastasis, and survival of tumors. Chronic lymphocytic leukemia (CLL) B cells of many patients can rapidly undergo apoptosis in vitro unless co-cultured with accessory cells, which elaborate factors that promote CLL-cell survival. A major type of such accessory cells is nurse-like cells (NLC), which prior studies demonstrated could differentiate from CD14+ blood mononuclear cells when exposed to CLL B cells in vitro, and presumably in vivo. In the absence of CLL cells, blood mononuclear CD14+ cells typically differentiate into macrophages that have a distinctive morphology from that of NLC. We found that, compared with macrophages, NLCs consistently expressed significantly higher levels of B-cell activating factor (BAFF) (n=11), a tumor-necrosis-factor (TNF)-related ligand that promotes survival of B cells, including CLL B cells. To investigate the influence of CLL cells on the differentiation of NLC in vitro, purified CLL B cells or B cells of healthy donors were cultured for 24h to generate conditioned media. When CD14+ blood mononuclear cells of healthy donors were cultured for 7 days in media conditioned by CLL cells, but not in non-conditioned media or in media conditioned by normal B cells, the cells assumed the morphology of NLC, acquired high-level expression of BAFF, and developed an enhanced capacity to support CLL cell survival in vitro. Similar activity was also found in the sera of CLL patients, but not in the sera of healthy control donors. The capacity of either CLL-conditioned media (n=4) or CLL-patient sera (n=9) to induce NLC differentiation leading to increased expression of BAFF was significantly reduced by a decoy receptor for TNF-a, suggesting that TNF-a contributes to the differentiation of NLC in vitro. However, TNF-a alone was not sufficient to induce high-level expression of BAFF on blood mononuclear CD14+ cells, suggesting that additional factors are involved. The results show a previously unrecognized symbiosis between CLL cells and accessory cells in their microenvironment. Because the survival of neoplastic B cells in vivo potentially depends upon NLC found in the lymphoid tissues of patients with CLL, identification of the factor(s) involved in inducing their differentiation could provide a novel target for therapy of patients with this disease.


2020 ◽  
Author(s):  
Haixiang Qin ◽  
Yang Yang ◽  
Bo Jiang ◽  
Chun Pan ◽  
Wei Chen ◽  
...  

Abstract Background Previous studies have demonstrated that transcription factor SOX9 which was reactivated in prostate cancer (Pca) and promoted tumor growth was a poor prognostic biomarker for Pca. Nevertheless, the regulatory mechanism underlying SOX9 upregulation in Pca still remains unclear. Several cytokines widely distributed in the tumor microenvironment (TME) have been reported to be involved in the regulation of SOX9, suggesting that cancer-associated fibroblasts (CAFs), one of the main sources of secreted factors in TME, may play a role in regulating SOX9 expression. Methods Herein, an in vitro model of paracrine interaction between primary CAFs and Pca cells (both AR-positive and AR-negative Pca cells), was applied to investigate the molecular mechanism of SOX9 upregulation during Pca progression. The regulatory axis was validated by in vivo experiments and The Cancer Genome Atlas (TCGA) data. Results Conditional medium from Pca CAFs (CAF-CM) upregulated the expression of SOX9, which was also proved to be essential for CAF-induced tumor progression. Further analysis showed that it was hepatocyte growth factor (HGF) secreted by CAFs that was responsible for the SOX9 elevation in Pca cells via activating c-Met signaling. Mechanistically, HGF/c-Met signaling specifically activated MEK1/2-ERK1/2 pathway which then induced phosphorylated status and protein upregulation of FRA1. Furthermore, ChIP assay demonstrated that FRA1 transcriptionally upregulated SOX9 expression by binding to the TPA-responsive element (TRE) sequence in the promoter of SOX9 gene. We also found that HGF/c-Met-ERK1/2-FRA1-SOX9 axis was relatively conserved in human and mouse species by validating in mouse Pca cells (RM-1). Conclusions Our results revealed a novel insight into the molecular mechanism that SOX9 expression in Pca cells is promoted by CAFs, through the HGF/cMet-ERK1/2-FRA1 axis. Besides, SOX9 may serve as an alternative marker for the activated HGF/c-Met signaling to enroll the optimal Pca patients for HGF/c-Met inhibition treatment, since it is much more stable and easier to detect.


2006 ◽  
Vol 110 (3) ◽  
pp. 279-291 ◽  
Author(s):  
Ingunn Holen ◽  
Claire M. Shipman

OPG (osteoprotegerin), a secreted member of the TNF (tumour necrosis factor) receptor superfamily, has a variety of biological functions which include the regulation of bone turnover. OPG is a potent inhibitor of osteoclastic bone resorption and has been investigated as a potential therapeutic for the treatment of both osteoporosis and tumour-induced bone disease. Indeed, in murine models of cancer-induced bone disease, inhibition of osteoclastic activity by OPG was also associated with a reduction in tumour burden. The discovery that OPG can bind to and inhibit the activity of TRAIL (TNF-related apoptosis-inducing ligand) triggered extensive research into the potential role of OPG in the regulation of tumour cell survival. A number of reports from studies using in vitro models have shown that OPG protects tumour cells from the effects of TRAIL, thereby possibly providing tumour cells that produce OPG with a survival advantage. However, the ability of OPG to act as a tumour cell survival factor remains to be verified using appropriate in vivo systems. A third area of interest has been the use of OPG as a prognostic marker in various cancer types, including myeloma, breast and prostate cancer. This review provides an overview of the role of OPG in cancer, both in cancer-induced bone disease and in tumour growth and survival.


2020 ◽  
Author(s):  
Diana Salikhova ◽  
Tatiana Bukharova ◽  
Elvira Cherkashova ◽  
Daria Namestnikova ◽  
Georgy Leonov ◽  
...  

Abstract Background: Stem cell secretomes hold great promise for regenerative medicine. This study is focused on the secretome-mediated neuroprotective effects of the human induced pluripotent stem cell-derived neuronal and glial progenitor cells. Therapeutic properties of the secretomes were assessed under conditions of the hypoxia-induced neuronal damage in vitro and in vivo. Methods: Secretory activity of the cultured neuronal and glial progenitor cells was analyzed by proteomic and immunosorbent-based approaches. Conditioned media collected from the cultures was tested for neuroprotective properties in vitro and in vivo.In vitro experiments involved exposure of SH-SY5Y cells to the conditioned media during the recovery from the cobalt chloride-induced hypoxia. Neuroprotective effects were assessed by cell survival and neurite outgrowth. Cell survival indicators included MTT and LDH tests, vital staining with propidium iodide and Hoechst 33342, and polymerase chain reaction assay for the expression of apoptosis-related genes. Neurite outgrowth was assessed by alterations in SH-SY5Y cell morphology and MAP2/GAP43 gene expression dynamics. In vivo experiments involved intra-arterial administration of the conditioned media to laboratory rats during the recovery from experimental ischemic stroke. Neuroprotective effects were assessed by overall survival, neurologic deficit and infarct volume dynamics, as well as by the end-point values of the apoptosis- and inflammation-related gene expression levels, the extent of microglia/macrophage infiltration, and the numbers of newly formed blood vessels in the affected area of the brain. Results: Secretomes of glial and neuronal progenitor cells partially overlapped, with specific proteins (found in secretome of one of the studied cultures and absent from the other) constituting, respectively, 31% and 45%. The glial progenitor cell-conditioned media showed higher content of neurotrophins (BDNF, GDNF, CNTF and NGF).Moreover, the glial progenitor cell-conditioned media was superior to the neuronal progenitor cell-conditioned media in facilitating neurite outgrowth and increasing SHSY-5Y cell survival after the cobalt dichloride-induced hypoxia. In addition, intra-arterial infusion of the glial progenitor cell-conditioned media to the animals after experimental ischemic stroke significantly enhanced functional recovery and promoted tissue repair at the site of brain damage, as indicated by reduced microglia/macrophage infiltration, decreased expression of pro-apoptotic gene Bax and pro-inflammatory cytokine gene Tnf, increased expression of anti-inflammatory cytokine genes (Il4, Il10, Il13), and increased numbers of newly formed blood vessels within the damaged area. None of these effects were exerted by the neuronal progenitor cell-conditioned media. Conclusions: The results indicate pronounced cytoprotective, anti-inflammatory and angionenic properties of soluble factors secreted by glial progenitor cells.


2021 ◽  
Author(s):  
Diana Salikhova ◽  
Tatiana Bukharova ◽  
Elvira Cherkashova ◽  
Daria Namestnikova ◽  
Georgy Leonov ◽  
...  

Abstract Background: Stem cell secretomes hold great promise for regenerative medicine. This study is focused on the secretome-mediated neuroprotective effects of the human induced pluripotent stem cell-derived neuronal and glial progenitor cells. Therapeutic properties of the secretomes were assessed under conditions of the hypoxia-induced neuronal damage in vitro and in vivo. Methods: Secretory activity of the cultured neuronal and glial progenitor cells was analyzed by proteomic and immunosorbent-based approaches. Conditioned media collected from the cultures was tested for neuroprotective properties in vitro and in vivo.In vitro experiments involved exposure of SH-SY5Y cells to the conditioned media during the recovery from the cobalt chloride-induced hypoxia. Neuroprotective effects were assessed by cell survival and neurite outgrowth. Cell survival indicators included MTT and LDH tests, vital staining with propidium iodide and Hoechst 33342, and polymerase chain reaction assay for the expression of apoptosis-related genes. Neurite outgrowth was assessed by alterations in SH-SY5Y cell morphology and MAP2/GAP43 gene expression dynamics. In vivo experiments involved intra-arterial administration of the conditioned media to laboratory rats during the recovery from experimental ischemic stroke. Neuroprotective effects were assessed by overall survival, neurologic deficit and infarct volume dynamics, as well as by the end-point values of the apoptosis- and inflammation-related gene expression levels, the extent of microglia/macrophage infiltration, and the numbers of newly formed blood vessels in the affected area of the brain. Results: Secretomes of glial and neuronal progenitor cells partially overlapped, with specific proteins (found in secretome of one of the studied cultures and absent from the other) constituting, respectively, 31% and 45%. The glial progenitor cell-conditioned media showed higher content of neurotrophins (BDNF, GDNF, CNTF and NGF).Moreover, the glial progenitor cell-conditioned media was superior to the neuronal progenitor cell-conditioned media in facilitating neurite outgrowth and increasing SHSY-5Y cell survival after the cobalt dichloride-induced hypoxia. In addition, intra-arterial infusion of the glial progenitor cell-conditioned media to the animals after experimental ischemic stroke significantly enhanced functional recovery and promoted tissue repair at the site of brain damage, as indicated by reduced microglia/macrophage infiltration, decreased expression of pro-apoptotic gene Bax and pro-inflammatory cytokine gene Tnf, increased expression of anti-inflammatory cytokine genes (Il4, Il10, Il13), and increased numbers of newly formed blood vessels within the damaged area. None of these effects were exerted by the neuronal progenitor cell-conditioned media. Conclusions: The results indicate pronounced cytoprotective, anti-inflammatory and angionenic properties of soluble factors secreted by glial progenitor cells.


Sign in / Sign up

Export Citation Format

Share Document