1
|
Zhuang M, Zhang W, Cheng N, Zhou L, Liu D, Yan H, Fang G, Heng BC, Sun Y, Tong G. Human umbilical cord mesenchymal stromal cells promote the regeneration of severe endometrial damage in a rat model. Acta Biochim Biophys Sin (Shanghai) 2021; 54:148-151. [PMID: 35130612 PMCID: PMC9909317 DOI: 10.3724/abbs.2021015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Affiliation(s)
- Mei Zhuang
- Department of GynecologyShanghai Traditional Chinese Medicine Integrated HospitalShanghai University of Traditional Chinese MedicineShanghai200082China
| | - Wuwen Zhang
- Reproductive Medicine CenterShuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghai201203China,Correspondence address. Tel: +86-21-20256688; E-mail: (W.Z.) / E-mail: (Y.S.) / E-mail: (G.T.)@
| | - Nuo Cheng
- Reproductive Medicine CenterShuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghai201203China
| | - Ling Zhou
- Department of Obstetrics and GynecologyStrategic Support Force Medical CenterBeijing100101China
| | - Dan Liu
- Reproductive Medicine CenterShuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghai201203China
| | - Hua Yan
- Reproductive Medicine CenterShuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghai201203China
| | - Ge Fang
- Reproductive Medicine CenterShuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghai201203China
| | - Boon Chin Heng
- Hospital & Institute of Obstetrics and GynecologyFudan University Shanghai Medical CollegeShanghai200011China
| | - Yan Sun
- Correspondence address. Tel: +86-21-20256688; E-mail: (W.Z.) / E-mail: (Y.S.) / E-mail: (G.T.)@
| | - Guoqing Tong
- Reproductive Medicine CenterShuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghai201203China,Correspondence address. Tel: +86-21-20256688; E-mail: (W.Z.) / E-mail: (Y.S.) / E-mail: (G.T.)@
| |
Collapse
|
2
|
Fibrin Clots Maintain the Viability and Proliferative Capacity of Human Mesenchymal Stem Cells: An In Vitro Study. Clin Orthop Relat Res 2020; 478:653-664. [PMID: 31842142 PMCID: PMC7145080 DOI: 10.1097/corr.0000000000001080] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Augmentation of soft-tissue repairs with an autologous fibrin clot has been used clinically for nearly four decades; however, fibrin clots tend to produce an abundance of scar tissue, which is known to inhibit soft-tissue regeneration. Mesenchymal stem cells (MSCs) embedded in fibrin clots before repair could reduce scar tissue deposition and facilitate soft-tissue regeneration. To our knowledge, no published studies have directly evaluated the viability or bioactivity of MSCs in fresh human fibrin clots over time. The purpose of this study was to evaluate the viability and bioactivity of human MSCs inside human fibrin clots over time in nutritive and non-nutritive culture media. QUESTIONS/PURPOSES We hypothesized that human MSCs would (1) be captured inside fibrin clots and retain their proliferative capacity, (2) remain viable for at least 7 days in the fibrin clots, (3) maintain their proliferative capacity for at least 7 days in the fibrin clots without evidence of active apoptosis, and (4) display similar viability and proliferative capacity when cultured in a non-nutritive medium over the same time periods. METHODS Twelve patients (mean age 33.7 years; range 4-72 years) who underwent elective knee surgery were approached between February 2016 and October 2017; all patients agreed to participate and were enrolled. MSCs isolated from human skeletal muscle and banked after prior studies were used for this analysis. On the day of surgery and after expansion of the MSC population, 3-mL aliquots of phosphate-buffered saline containing approximately 600,000 labeled with anti-green fluorescent protein (GFP) antibodies were transported to the operating room, mixed in 30 mL of venous blood from each enrolled patient, and stirred at 95 rpm for 10 minutes to create MSC-embedded fibrin clots. The fibrin clots were transported to the laboratory with their residual blood for analysis. Eleven samples were analyzed after exclusion of one sample because of a processing error. MSC capture was qualitatively demonstrated by enzymatically digesting half of each clot specimen, thus releasing GFP-positive MSCs into culture. The released MSCs were allowed to culture for 7 days. Manual counting of GFP-positive MSCs was performed at 2, 3, 4, and 7 days using an inverted microscope at 100 x magnification to document the change in the number of GFP-positive MSCs over time. The intact remaining half of each clot specimen was immediately placed in proliferation media and allowed to culture for 7 days. On Days 1, 2, 3, 4, and 7, a small portion of the clot was excised, flash-frozen, cryosectioned (8-μm thickness), and immunostained with antibodies specific to GFP, Ki67 (indicative of active proliferation), and cleaved caspase-3 ([CC3]; indicative of active apoptosis). Using an inverted microscope, we obtained MSC cell counts manually at time zero and after 1, 2, 3, 4, and 7 days of culture. Intact fresh clot specimens were immediately divided in half; one half was placed in nutritive (proliferation media) and the other was placed in non-nutritive (saline) media for 1, 2, 3, 4, and 7 days. At each timepoint, specimens were processed in an identical manner as described above, and a portion of each clot specimen was excised, immediately flash-frozen with liquid nitrogen, cryosectioned (8-μm thickness), and visualized at 200 x using an inverted microscope. The numbers of stain-positive MSCs per field of view, per culture condition, per timepoint, and per antibody stain type were counted manually for a quantitative analysis. Raw data were statistically compared using t-tests, and time-based correlations were assessed using Pearson's correlation coefficients. Two-tailed p values of less than 0.05 (assuming unequal variance) were considered statistically significant. RESULTS Green fluorescence, indicative of viable GFP-positive MSCs, was absent in all residual blood samples after 48 hours of culturing; GFP-positive MSCs were visualized after enzymatic digestion of clot matrices. The number of GFP-positive MSCs per field of view increased between the 2-day and 7-day timepoints (mean 5.4 ± 1.5; 95% confidence interval, 4.7-6.1 versus mean 17.0 ± 13.6; 95% CI, 10.4-23.5, respectively; p = 0.029). Viable GFP-positive MSCs were present in each clot cryosection at each timepoint up to 7 days of culturing (mean 6.2 ± 4.3; 95% CI, 5.8-6.6). There were no differences in MSC counts between any of the timepoints. There was no visible evidence of GFP +/CC3 + double-positive MSCs. Combining all timepoints, there were 0.34 ± 0.70 (95% CI, 0.25-0.43) GFP+/Ki67+ double-positive MSCs per field of view. The mitotic indices at time zero and Day 7 were 7.5% ± 13.4% (95% CI, 3.0%-12.0%) and 7.2% ± 14.3% (95% CI, 3.3%-12,1%), respectively (p = 0.923). There was no visible evidence of GFP +/CC3 + double-positive MSCs (active apoptosis) at any timepoint. For active proliferation in saline-cultured fibrin clots, we found averages of 0.1 ± 0.3 (95% CI, 0.0-0.2) and 0.4 ± 0.9 (95% CI, 0.0-0.8) GFP/Ki67 double-positive MSCs at time zero and Day 7, respectively (p = 0.499). The mitotic indices in saline culture at time zero and Day 7 were 2.9% ± 8.4% (95% CI, 0.0%-5.8%) and 9.1% ± 20.7% (95% CI, 1.2%-17.0%; p = 0.144). There was no visible evidence of GFP +/CC3 + double-positive MSCs (active apoptosis) at any timepoint in either culturing condition. CONCLUSION These preliminary in vitro results show that human MSCs mixed in unclotted fresh human venous blood were nearly completely captured in fibrin clots and that seeded MSCs were capable of maintaining their viability, proliferation capacity, and osteogenic differentiation capacity in the fibrin clot for up to 7 days, independent of external sources of nutrition. CLINICAL RELEVANCE Fresh human fibrin clots have been used clinically for more than 30 years to improve soft-tissue healing, albeit with scar tissue. Our results demonstrate that allogenic human MSCs, which reduce soft-tissue scarring, can be captured and remain active inside human fibrin clots, even in the absence a nutritive culture medium.
Collapse
|
3
|
Mizukami A, Thomé CH, Ferreira GA, Lanfredi GP, Covas DT, Pitteri SJ, Swiech K, Faça VM. Proteomic Identification and Time-Course Monitoring of Secreted Proteins During Expansion of Human Mesenchymal Stem/Stromal in Stirred-Tank Bioreactor. Front Bioeng Biotechnol 2019; 7:154. [PMID: 31297369 PMCID: PMC6607109 DOI: 10.3389/fbioe.2019.00154] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 06/10/2019] [Indexed: 12/14/2022] Open
Abstract
The therapeutic potential of mesenchymal stem/stromal cells (MSC) is widely recognized for the treatment of several diseases, including acute graft-vs.-host disease (GVHD), hematological malignancies, cardiovascular, bone, and cartilage diseases. More recently, this therapeutic efficacy has been attributed to the bioactive molecules that these cells secrete (secretome), now being referred as medicinal signaling cells. This fact raises the opportunity of therapeutically using MSC-derived soluble factors rather than cells themselves, enabling their translation into the clinic. Indeed, many clinical trials are now studying the effects of MSC-secretome in the context of cell-free therapy. MSC secretome profile varies between donors, source, and culture conditions, making their therapeutic use very challenging. Therefore, identifying these soluble proteins and evaluating their production in a reproducible and scalable manner is even more relevant. In this work, we analyzed the global profile of proteins secreted by umbilical cord matrix (UCM) derived-MSC in static conditions by using mass spectrometry, enabling the identification of thousands of proteins. Afterwards, relevant proteins were chosen and monitored in the supernatant of a fully-controllable, closed and scalable system (bioreactor) by using multiple reaction monitoring (MRM) mass spectrometric technique in a time-dependent manner. The results showed that the majority of interesting proteins were enriched through time in culture, with the last day of culture being the ideal time for supernatant collection. The use of this regenerative "soup," which is frequently discarded, could represent a step toward a safe, robust and reproducible cell-free product to be used in the medical therapeutic field. The future use of chemically defined culture-media will certainly facilitate secretome production according to Good Manufacturing Practice (GMP) standards.
Collapse
Affiliation(s)
- Amanda Mizukami
- Faculty of Medicine of Ribeirão Preto, Hemotherapy Center of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Carolina Hassibe Thomé
- Faculty of Medicine of Ribeirão Preto, Hemotherapy Center of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil.,Department of Biochemistry and Immunology, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Germano Aguiar Ferreira
- Faculty of Medicine of Ribeirão Preto, Hemotherapy Center of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil.,Department of Biochemistry and Immunology, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Guilherme Pauperio Lanfredi
- Department of Biochemistry and Immunology, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Dimas Tadeu Covas
- Faculty of Medicine of Ribeirão Preto, Hemotherapy Center of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Sharon J Pitteri
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, Stanford, CA, United States
| | - Kamilla Swiech
- Faculty of Medicine of Ribeirão Preto, Hemotherapy Center of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil.,Department of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences of Ribeirao Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Vitor Marcel Faça
- Faculty of Medicine of Ribeirão Preto, Hemotherapy Center of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil.,Department of Biochemistry and Immunology, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
4
|
Polymorphisms in the 3'-UTR of SCD5 gene are associated with hepatocellular carcinoma in Korean population. Mol Biol Rep 2018; 45:1705-1714. [PMID: 30168096 DOI: 10.1007/s11033-018-4313-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 08/17/2018] [Indexed: 12/28/2022]
Abstract
The purpose of the study was to assess the relationship between polymorphisms of the SCD5 and MMP1 gene and hepatocellular carcinoma (HCC). The gene polymorphisms with a minor allele frequency (MAF) > 0.05 were selected eight SNPs (rs6840, rs1065403, rs3821974, and rs3733230 in 3'-UTR; rs4693472, rs3733227, rs1848067, and rs6535374 in intron region) of SCD5 gene and two SNPs (rs1799750 and rs1144393 in promoter region) of MMP1 gene. The genotype of SCD5 and MMP1 gene SNPs were determined by direct sequencing and pyrosequencing, respectively. One hundred sixty-two patients with HCC and two hundred twenty-five control subjects were recruited in Korean male population. In terms of genotype frequencies, SCD5 genotype TC, GA, AG, and CG of rs6840, rs1065403, rs3821974, and rs3733230, respectively were higher in control group than HCC. In addition, these genotype decreased the risk (rs6840; OR 0.55, 95% CI 0.31-0.99; rs1065403; OR 0.46, 95% CI 0.26-0.83; rs3821974; OR 0.56, 95% CI 0.31-0.99; rs3733230; OR 0.62, 95% CI 0.34-1.12) of HCC, which may work as a prevention of HCC. At least one minor allele carrier of SCD5 gene polymorphisms were related to decreased risk of HCC for AFP cut-point levels > 200 or > 400 ng/ml, respectively. Our results indicate that polymorphisms in the 3'-UTR of the SCD5 gene may associated with HCC in the Korean male population.
Collapse
|
5
|
Daviran M, Longwill SM, Casella JF, Schultz KM. Rheological characterization of dynamic remodeling of the pericellular region by human mesenchymal stem cell-secreted enzymes in well-defined synthetic hydrogel scaffolds. SOFT MATTER 2018; 14:3078-3089. [PMID: 29667686 PMCID: PMC5928794 DOI: 10.1039/c8sm00408k] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Human mesenchymal stem cells (hMSCs) dynamically remodel their microenvironment during basic processes, such as migration and differentiation. Migration requires extracellular matrix invasion, necessitating dynamic cell-material interactions. Understanding these interactions is critical to advancing materials designs that harness and manipulate these processes for applications including wound healing and tissue regeneration. In this work, we encapsulate hMSCs in a cell-degradable poly(ethylene glycol)-peptide hydrogel to determine how cell-secreted enzymes, specifically matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs), create unique pericellular microenvironments. Using multiple particle tracking microrheology (MPT), we characterize spatio-temporal rheological properties in the pericellular region during cell-mediated remodeling. In MPT, the thermal motion of probes embedded in the network is measured. A newly designed sample chamber that limits probe drift during degradation and minimizes high value antibody volumes required for cell treatments enables MPT characterization. Previous MPT measurements around hMSCs show that directly around the cell the scaffold remains intact with the cross-link density decreasing as distance from the cell increases. This degradation profile suggests that hMSCs are simultaneously secreting TIMPs, which are inactivating MMPs through MMP-TIMP complexes. By neutralizing TIMPs using antibodies, we characterize the changes in matrix degradation. TIMP inhibited hMSCs create a reaction-diffusion type degradation profile where MMPs are actively degrading the matrix immediately after secretion. In this profile, the cross-link density increases with increasing distance from the cell. This change in material properties also increases the speed of migration. This simple treatment could increase delivery of hMSCs to injuries to aid wound healing and tissue regeneration.
Collapse
Affiliation(s)
- Maryam Daviran
- Department of Chemical and Biomolecular Engineering, Lehigh University, 111 Research Dr., Iacocca Hall, Bethlehem, PA 18015, USA.
| | | | | | | |
Collapse
|
6
|
Lai YL, Gong CL, Fu CK, Yueh TC, Tsai CW, Chang WS, Hsiao CL, Yen ST, Li HT, Jeng LB, Wang SC, Bau DAT. The Contribution of Matrix Metalloproteinase-1 Genotypes to Hepatocellular Carcinoma Susceptibility in Taiwan. Cancer Genomics Proteomics 2017; 14:119-125. [PMID: 28387651 DOI: 10.21873/cgp.20024] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 02/28/2017] [Accepted: 02/28/2017] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND/AIM Metalloproteinases (MMPs) are a family of proteases which have been shown to be overexpressed in various types of cancers. However, the contribution of MMP1 genotype to hepatocellular carcinoma (HCC) has not been well studied. This study aimed to evaluate the contribution of MMP1 promoter 1607 genotype to the risk of HCC in Taiwan, where HCC incidence is relatively high in the world. MATERIALS AND METHODS In this case-control study, MMP1 genotype and its interaction with consumption of cigarettes and alcohol in determining HCC risk was investigated among 298 HCC patients and 889 age- and gender-matched healthy controls. RESULTS The percentages of ever smokers and ever alcohol drinkers were much higher in the case group than in the control group. The percentages of 2G/2G, 1G/2G and 1G/1G for MMP1 promoter 1607 genotype were 37.2%, 38.3% and 24.5% in the HCC group and 34.8%, 44.0% and 21.2% in the control group, respectively (p for trend=0.2048). The allelic frequency distribution analysis showed the variant 1G allele of MMP1 promoter 1607 conferred similar HCC susceptibility as the wild-type 2G allele (odds ratio (OR)=1.01, 95% confidence interval (CI)=0.84-1.22, p=0.8735). As for the gene-lifestyle interaction, there was an obvious protective effect of MMP1 promoter 1607 1G allele on the risk of HCC among non-smokers, but not non-smokers, even alcohol drinkers or non-drinkers. CONCLUSION The 1G allele of MMP1 promoter 1607 may have a protective effect on HCC risk for non-smokers in Taiwan and further validations are needed in other population groups.
Collapse
Affiliation(s)
- Yi-Liang Lai
- Taichung Armed Forces General Hospital, Taichung, Taiwan, R.O.C
| | - Chi-Li Gong
- Department of Physiology, China Medical University, Taichung, Taiwan, R.O.C
| | - Chun-Kai Fu
- Taichung Armed Forces General Hospital, Taichung, Taiwan, R.O.C.,Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, R.O.C
| | - Te-Cheng Yueh
- Taichung Armed Forces General Hospital, Taichung, Taiwan, R.O.C.,Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, R.O.C
| | - Chia-Wen Tsai
- Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Wen-Shin Chang
- Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Chieh-Lun Hsiao
- Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Shiou-Ting Yen
- Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Hsin-Ting Li
- Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Long-Bin Jeng
- Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Shou-Cheng Wang
- Taichung Armed Forces General Hospital, Taichung, Taiwan, R.O.C. .,Natiosnal Defense Medical Center, Taipei, Taiwan, R.O.C
| | - DA-Tian Bau
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, R.O.C. .,Terry Fox Cancer Research Laboratory, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan, R.O.C.,Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan, R.O.C
| |
Collapse
|
7
|
Extracts from Curcuma zedoaria Inhibit Proliferation of Human Breast Cancer Cell MDA-MB-231 In Vitro. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2014; 2014:730678. [PMID: 24883070 PMCID: PMC4026840 DOI: 10.1155/2014/730678] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 04/03/2014] [Accepted: 04/07/2014] [Indexed: 12/19/2022]
Abstract
Objective. To evaluate the effect of petroleum ether extracts of Curcuma zedoaria on the proliferation of human triple negative breast cancer cell line MDA-MB-231. Methods. The reagents were isolated from Curcuma zedoaria by petroleum ether fraction. It was assayed by CCK8 for MDA-MB-231 cellular viability with various concentrations and days, cell cycle analyses, Western Blot analysis, and Realtime Reverse Transcriptase PCR analyses for chemokines molecules including E-cadherin, and E-selectin, and adhesion molecules including CCR7, SLC, SDF-1, and CXCR4. Epirubicin was used as control in the study. Results. MDA-MB-231 cells were inhibited by petroleum ether extracts of Curcuma zedoaria (P < 0.05), and the inhibition rate was dependent on concentrations and time. Petroleum ether extracts of Curcuma zedoaria as well as Epirubicin produce a significant G0/G1 cell cycle arrest. The level of expression of proteins E-cadherin and E-cadherin mRNA was significantly increased, while proteins SDF-1, CCR7, and CXCR4 mRNA were decreased after being incubated with petroleum ether extracts of Curcuma zedoaria at the concentrations of 300 μg/mL than control (P < 0.05). The differences were that the protein CXCR4 mRNA expression level was higher than vehicle. Conclusions. MDA-MB-231 cells were inhibited by petroleum ether extracts of Curcuma zedoaria.
Collapse
|
8
|
Jin Y, Roycik MD, Bosco DB, Cao Q, Constantino MH, Schwartz MA, Sang QXA. Matrix metalloproteinase inhibitors based on the 3-mercaptopyrrolidine core. J Med Chem 2013; 56:4357-73. [PMID: 23631440 DOI: 10.1021/jm400529f] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
New series of pyrrolidine mercaptosulfide, 2-mercaptocyclopentane arylsulfonamide, and 3-mercapto-4-arylsulfonamidopyrrolidine matrix metalloproteinase inhibitors (MMPIs) were designed, synthesized, and evaluated. Exhibiting unique properties over other MMPIs (e.g., hydroxamates), these newly reported compounds are capable of modulating activities of several MMPs in the low nanomolar range, including MMP-2 (~2 to 50 nM), MMP-13 (~2 to 50 nM), and MMP-14 (~4 to 60 nM). Additionally these compounds are selective to intermediate- and deep-pocket MMPs but not shallow-pocketed MMPs (e.g., MMP-1, ~850 to >50,000 nM; MMP-7, ~4000 to >25,000 nM). Our previous work with the mercaptosulfide functionality attached to both cyclopentane and pyrrolidine frameworks demonstrated that the cis-(3S,4R)-stereochemistry was optimal for all of the MMPs tested. However, in our newest compounds an interesting shift of preference to the trans form of the mercaptosulfonamides was observed with increased oxidative stability and biological compatibility. We also report several kinetic and biological characteristics showing that these compounds may be used to probe the mechanistic activities of MMPs in disease.
Collapse
Affiliation(s)
- Yonghao Jin
- Department of Chemistry and Biochemistry and Institute of Molecular Biophysics, Florida State University, Tallahassee, Florida, USA
| | | | | | | | | | | | | |
Collapse
|
9
|
Ling X, Spaeth E, Chen Y, Shi Y, Zhang W, Schober W, Hail N, Konopleva M, Andreeff M. The CXCR4 antagonist AMD3465 regulates oncogenic signaling and invasiveness in vitro and prevents breast cancer growth and metastasis in vivo. PLoS One 2013; 8:e58426. [PMID: 23484027 PMCID: PMC3590173 DOI: 10.1371/journal.pone.0058426] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Accepted: 02/05/2013] [Indexed: 01/12/2023] Open
Abstract
CXCR4, the receptor for stromal-derived factor-1, is reportedly involved in breast carcinogenesis. However, the mechanisms through which CXCR4 contributes to breast cancer cell growth and metastases are poorly understood. In this study, we examined the putative in vitro and in vivo anti-cancer effects of the specific CXCR4 inhibitor AMD3465. Here, we report that AMD3465 triggers a reduction in breast cancer cell invasiveness in vitro, and promotes marked changes in oncogenic signaling proteins including a reduction in STAT3, JAK2, AKT, and CXCR4 phosphorylation and the reduced expression of GSK3 and cMYC. Using three breast cancer cell lines as murine syngeneic immunocompetent breast cancer models, we found that AMD3465 inhibited breast tumor formation and reduced tumor cell metastases to the lung and liver. Furthermore, treatment with AMD3465 significantly reduced the infiltration of myeloid CD11b positive cells at the aforementioned metastatic sites as well as the spleen implying this agent could regulate the formation of the tumor microenvironment and conceivably the premetastatic niche. In conclusion, our studies suggest that AMD3465 inhibits breast cancer growth and metastases by acting on tumor cells as well as immune cells that constitute the tumor microenvironment. This process appears to be regulated, at least in part, through the modulation of oncogenic signaling that includes the STAT3 pathway. Thus, CXCR4 could be a novel target for breast cancer therapy.
Collapse
Affiliation(s)
- Xiaoyang Ling
- Department of Leukemia, University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Erika Spaeth
- Department of Leukemia, University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Ye Chen
- Department of Leukemia, University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Yuexi Shi
- Department of Leukemia, University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Weiguo Zhang
- Department of Leukemia, University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Wendy Schober
- Department of Leukemia, University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Numsen Hail
- Department of Leukemia, University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Marina Konopleva
- Department of Leukemia, University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
| | - Michael Andreeff
- Department of Leukemia, University of Texas M. D. Anderson Cancer Center, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
10
|
Huang YC, Leung VYL, Lu WW, Luk KDK. The effects of microenvironment in mesenchymal stem cell-based regeneration of intervertebral disc. Spine J 2013; 13:352-62. [PMID: 23340343 DOI: 10.1016/j.spinee.2012.12.005] [Citation(s) in RCA: 134] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2011] [Revised: 07/08/2012] [Accepted: 12/09/2012] [Indexed: 02/03/2023]
Abstract
BACKGROUND CONTEXT Recent studies have demonstrated new therapeutic strategy using transplantation of mesenchymal stem cells (MSCs), especially bone marrow-derived MSCs (BM-MSCs), to preserve intervertebral disc (IVD) structure and functions. It is important to understand whether and how the MSCs survive and thrive in the hostile microenvironment of the degenerated IVD. Therefore, this review majorly examines how resident disc cells, hypoxia, low nutrition, acidic pH, mechanical loading, endogenous proteinases, and cytokines regulate the behavior of the exogenous MSCs. PURPOSE To review and summarize the effect of the microenvironment in biological characteristics of BM-MSCs for IVD regeneration; the presence of endogenous stem cells and the state of the art in the use of BM-MSCs to regenerate the IVD in vivo were also discussed. STUDY DESIGN Literature review. METHODS MEDLINE electronic database was used to search for articles concerning stem/progenitor cell isolation from the IVD, regulation of the components of microenvironment for MSCs, and MSC-based therapy for IVD degeneration. The search was limited to English language. RESULTS Stem cells are probably resident in the disc, but exogenous stem cells, especially BM-MSCs, are currently the most popular graft cells for IVD regeneration. The endogenous disc cells and the biochemical and biophysical components in the degenerating disc present a complicated microenvironment to regulate the transplanted BM-MSCs. Although MSCs regenerate the mildly degenerative disc effectively in the experimental and clinical trials, many underlying questions are in need of further investigation. CONCLUSIONS There has been a dramatic improvement in the understanding of potential MSC-based therapy for IVD regeneration. The use of MSCs for IVD degeneration is still at the stage of preclinical and Phase 1 studies. The effects of the disc microenvironment in MSCs survival and function should be closely studied for transferring MSC transplantation from bench to bedside successfully.
Collapse
Affiliation(s)
- Yong-Can Huang
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, Queen Mary Hospital, The University of Hong Kong, 5/F Professor Block, Pokfulam, Hong Kong, China
| | | | | | | |
Collapse
|
11
|
Jiang L, Ma A, Song L, Hu Y, Dun H, Daloze P, Yu Y, Jiang J, Zafarullah M, Chen H. Cartilage regeneration by selected chondrogenic clonal mesenchymal stem cells in the collagenase-induced monkey osteoarthritis model. J Tissue Eng Regen Med 2013; 8:896-905. [PMID: 23335439 DOI: 10.1002/term.1676] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Revised: 09/11/2012] [Accepted: 11/05/2012] [Indexed: 12/29/2022]
Abstract
Osteoarthritis (OA) is the most common form of arthritis, in which cartilage is irreversibly degraded, causing severe pain and disability. Current therapeutic strategies cannot repair damaged cartilage. We evaluated the repair potential of selected chondrogenic clonal MSCs (sC-MSCs) by delivering them into the injured cartilage site in a collagenase-induced OA model in Cynomolgus monkeys. In vitro characterization showed that the isolated monkey sC-MSCs and polyclonal MSCs (P-MSCs) expressed mesenchymal stem cell markers and could differentiate into chondrocytes. The articular cartilage lesions in animals were treated with normal saline (NS), autologous P-MSCs and sC-MSCs, respectively, by direct delivery. The clinical parameters, radiographic images, histological and immunohistochemical examinations at weeks 8, 16 and 24 post-treatment demonstrated that the abrasions of articular cartilage were significantly improved and repaired by MSC-based treatment, particularly in the sC-MSC-treated group, which displayed consistently higher histological scores than those of other groups. In summary, treatment with sC-MSCs can effectively improve the healing of cartilage lesions in the Cynomolgus monkey collagenase-induced OA model. Due to the genetic proximity of monkey and human, the therapeutic strategy presented in this study will have broad applications in clinical practice.
Collapse
Affiliation(s)
- Li Jiang
- Department of Surgery, CRCHUM, Notre Dame Hospital, University of Montreal, Quebec, Canada; Department of Orthopaedics, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Faça VM. Human mesenchymal stromal cell proteomics: contribution for identification of new markers and targets for medicine intervention. Expert Rev Proteomics 2012; 9:217-30. [PMID: 22462791 DOI: 10.1586/epr.12.9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Mesenchymal stem or stromal cells (MSCs) have become of great interest for cell-based therapy owing to their roles in tissue repair and immune suppression. MSCs have the ability to differentiate into specialized tissues, including bone, cartilage and muscle, among several others. Furthermore, it has been found that MSCs can also serve as cellular factories that secrete mediators to stimulate in situ regeneration of injured tissues. Proteomics has contributed significantly to the identification of new proteins to improve cellular characterization of MSCs, to identify new targets for therapeutic intervention and to elucidate important pathways utilized by MSCs to differentiate into distinct tissues. As proteomics technology advances, several studies can be revisited and analyzed in depth, employing state-of-the-art approaches, helping to uncover the cellular mechanisms utilized by MSCs to exert their regenerative functionalities. In this article, we will review the progress made so far and discuss further opportunities for proteomics to contribute to the clinical applications of MSCs.
Collapse
Affiliation(s)
- Vitor Marcel Faça
- Department of Biochemistry & Immunology, Faculty of Medicine of Ribeirão Preto, University of São Paulo, Brazil.
| |
Collapse
|
13
|
Bačenková D, Rosocha J, Tóthová T, Rosocha L, Šarisský M. Isolation and basic characterization of human term amnion and chorion mesenchymal stromal cells. Cytotherapy 2012; 13:1047-56. [PMID: 21916779 DOI: 10.3109/14653249.2011.592522] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND AIMS Emerging evidence suggests human placental membrane is a valuable source of mesenchymal stromal cells (MSC). Amnion and chorion are tissues of early embryologic origin that may entail progenitor potential. These tissues are abundantly available and ethically unobjectionable and, because they are discarded post-partum, they can be widely used for extensive research and eventually for therapeutic studies. METHODS We looked at the cells isolated from the six amnions and chorions of term placentas of gestational weeks 39 ± 1. Isolated cells were characterized by morphologic and immunophenotypic analysis. RESULTS With flow cytometry immunophenotype analysis, amnion- and chorion-derived cells were positive for MSC markers, and negative for hematopoietic markers. Immunocytochemical staining was positive for the embryonic cell markers Oct-3/4 and Rex-1. Oct-3/4 is a POU transcription factor that is expressed in embryonic stem (ES) cells and germ cells, and its expression is required to sustain cell self-renewal and pluripotency. Oct-3/4 is the most recognized marker for totipotent ES cells. Rex-1 is a zinc finger family transcription factor that is highly expressed in embryonic stem cells. It is one of several gene markers used to identify undifferentiated stem cells, and its expression is downregulated upon stem cell differentiation. Amnion- and chorion-derived cells were capable, under differentiation conditions, to differentiate into to mesoderm lineages. CONCLUSIONS Phenotypic studies indicate MSC-like profiles in both amnion- and chorion-derived cells. Cells in vitro had fibroblastoid morphology. The in vitro growth behavior of such placenta-derived progenitor cells was similar to that of bone marrow MSC. Our results indicate that MSC can be easily isolated from the human term placenta. The human amniotic and chorion MSC maintained a marker profile similar to the mesenchymal progenitors and could be used for studies as an alternative source of MSC for further application in cellular therapy.
Collapse
Affiliation(s)
- Darina Bačenková
- Tissue Bank, PJ Šafárik University, Trieda SNP 1, Košice, Slovakia
| | | | | | | | | |
Collapse
|
14
|
Song C, Li G. CXCR4 and matrix metalloproteinase-2 are involved in mesenchymal stromal cell homing and engraftment to tumors. Cytotherapy 2010; 13:549-61. [PMID: 21171825 DOI: 10.3109/14653249.2010.542457] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND AIMS Bone marrow-derived mesenchymal stromal cells (BMSC) have been shown to migrate to injury, ischemia and tumor microenvironments. The mechanisms by which mesenchymal stromal cells (MSC) migrate across endothelium and home to the target tissues are not yet fully understood. METHODS We used rat BMSC to investigate the molecular mechanisms involved in their tropism to tumors in vitro and in vivo. RESULTS BMSC were shown to migrate toward four different tumor cells in vitro, and home to both subcutaneous and lung metastatic prostate tumor models in vivo. Gene expression profiles of MSC exposed to conditioned medium (CM) of various tumor cells were compared and revealed that matrix metalloproteinase-2 (MMP-2) expression in BMSC was downregulated after 24 h exposure to tumor CM. Chemokine (C-X-C motif) Receptor 4 (CXCR4) upregulation was also found in BMSC after 24 h exposure to tumor CM. Exposure to tumor cell CM enhanced migration of BMSC toward tumor cells. Stromal Cell-Derived Factor (SDF-1) inhibitor AMD3100 and MMP-2 inhibitor partly abolished the BMSC migration toward tumor cells in vitro. CONCLUSIONS These results suggest that the CXCR4 and MMP-2 are involved in the multistep migration processes of BMSC tropism to tumors.
Collapse
Affiliation(s)
- Chao Song
- Stem Cell and Regeneration Program, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, SAR, PR China
| | | |
Collapse
|
15
|
Mesenchymale Stammzellen für das „tissue engineering“ des Knochens. DER PATHOLOGE 2010; 31 Suppl 2:138-46. [DOI: 10.1007/s00292-010-1329-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
16
|
Marrow Stromal Cells for Cell-Based Therapy: The Role of Antiinflammatory Cytokines in Cellular Cardiomyoplasty. Ann Thorac Surg 2010; 90:190-7. [DOI: 10.1016/j.athoracsur.2010.02.074] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2009] [Revised: 02/23/2010] [Accepted: 02/26/2010] [Indexed: 11/24/2022]
|
17
|
Abstract
Cartilage repair is a very successful pioneering area of regenerative medicine in which techniques of in situ regeneration and cell and tissue transplantation dominate over cell-free approaches to generate durable neocartilage. This review concentrates on advantages and limitations of mesenchymal stem cell (MSC)-based cartilage repair strategies induced by marrow stimulation. Detailed knowledge on the biology of MSC will be discussed in light of the requirements for MSC recruitment, retention, proliferation and chondrogenic differentiation. An improved microenvironment with timely correlated signals from biomaterials, growth factors, proteases, adjacent cartilage and subchondral bone may be key to a third generation of techniques to regenerate hyaline cartilage.
Collapse
Affiliation(s)
- W Richter
- Department of Experimental Orthopaedics, Orthopaedic University Hospital, Heidelberg, Germany.
| |
Collapse
|
18
|
Schubert T, Kaufmann S, Wenke AK, Grässel S, Bosserhoff AK. Role of deleted in colon carcinoma in osteoarthritis and in chondrocyte migration. Rheumatology (Oxford) 2009; 48:1435-41. [PMID: 19745029 DOI: 10.1093/rheumatology/kep245] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
OBJECTIVE The concept of the chondrocyte as a stationary cell surrounded by an apparently impenetrable matrix has been challenged by in vitro observations in recent years. Chondrocyte migration may have a role in remodelling of the cartilage and pathological conditions. Candidate molecules are repellent factors for the regulation of chondrocyte migration, which are expressed in fetal and adult cartilage. We analysed the potential role of the receptor deleted in colon carcinoma (DCC) in chondrocytes, as this may exert attractive activities. METHODS Gene expression was determined by quantitative RT-PCR and immunohistochemistry, and gene regulation by electro mobility shift assay and chromatin immunoprecipitation. Functional assays on migration and differentiation were done after cell treatment and transfection. RESULTS DCC was shown to be specifically up-regulated in OA compared with normal chondrocytes in vitro and in vivo. Promoter analysis and transfection studies showed that the up-regulation of DCC in OA chondrocytes may be mediated by the transcription factors Sox9 and AP-2. Netrin-1, the ligand of DCC, was revealed to induce the migration of OA chondrocytes specifically. Expression of DCC in healthy chondrocytes by transient transfection significantly induced cell migration and chemotaxis to Netrin-1. DCC expression had no influence on cell differentiation; however, induction of MMP1 and -3 expression was observed. CONCLUSION Strong differential expression of DCC in OA compared with normal chondrocytes hints of a possible role of DCC in the pathophysiology of OA. The strong impact of the DCC receptor on cellular mobility of chondrocytes in vitro suggests a major relevance of migratory activities in physiological and pathological conditions of cartilage. However, definite proof of chondrocyte movements in vivo still has to be established.
Collapse
Affiliation(s)
- Thomas Schubert
- University of Regensburg, Institute of Pathology, Franz-Josef-Strauss-Allee 11, 93053 Regensburg, Germany
| | | | | | | | | |
Collapse
|
19
|
Heng BC, Cowan CM, Basu S. Comparison of enzymatic and non-enzymatic means of dissociating adherent monolayers of mesenchymal stem cells. Biol Proced Online 2009; 11:161-9. [PMID: 19495917 PMCID: PMC3055293 DOI: 10.1007/s12575-009-9001-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2009] [Accepted: 01/30/2009] [Indexed: 12/24/2022] Open
Abstract
The dissociation of adherent mesenchymal stem cell (MSC) monolayers with trypsin and enzyme-free dissociation buffer was compared. A significantly lower proportion of viable cells were obtained with enzyme-free dissociation buffers compared to trypsin. Subsequently, the dissociated cells were re-seeded on new cell culture dishes and were subjected to the MTT assay 24 h later. The proportion of viable cells that reattached was significantly lower for cells obtained by dissociation with enzyme-free dissociation buffer compared to trypsin. Frozen–thawed MSC displayed a similar trend, yielding consistently higher cell viability and reattachment rates when dissociated with trypsin compared to enzyme-free dissociation buffer. It was also demonstrated that exposure of trypsin-dissociated MSC to enzyme-free dissociation buffer for 1 h had no significant detrimental effect on cell viability.
Collapse
|
20
|
Kafadar KA, Yi L, Ahmad Y, So L, Rossi F, Pavlath GK. Sca-1 expression is required for efficient remodeling of the extracellular matrix during skeletal muscle regeneration. Dev Biol 2008; 326:47-59. [PMID: 19059231 DOI: 10.1016/j.ydbio.2008.10.036] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2008] [Revised: 09/16/2008] [Accepted: 10/20/2008] [Indexed: 01/03/2023]
Abstract
Sca-1 (Stem Cell Antigen-1) is a member of the Ly-6 family proteins that functions in cell growth, differentiation, and self-renewal in multiple tissues. In skeletal muscle Sca-1 negatively regulates myoblast proliferation and differentiation, and may function in the maintenance of progenitor cells. We investigated the role of Sca-1 in skeletal muscle regeneration and show here that Sca-1 expression is upregulated in a subset of myogenic cells upon muscle injury. We demonstrate that extract from crushed muscle upregulates Sca-1 expression in myoblasts in vitro, and that this effect is reversible and independent of cell proliferation. Sca-1(-/-) mice exhibit defects in muscle regeneration, with the development of fibrosis following injury. Sca-1(-/-) muscle displays reduced activity of matrix metalloproteinases, critical regulators of extracellular matrix remodeling. Interestingly, we show that the number of satellite cells is similar in wild-type and Sca-1(-/-) muscle, suggesting that in satellite cells Sca-1 does not play a role in self-renewal. We hypothesize that Sca-1 upregulates, directly or indirectly, the activity of matrix metalloproteinases, leading to matrix breakdown and efficient muscle regeneration. Further elucidation of the role of Sca-1 in matrix remodeling may aid in the development of novel therapeutic strategies for the treatment of fibrotic diseases.
Collapse
|
21
|
Lin J, Lindsey ML, Zhu B, Agrawal CM, Bailey SR. Effects of surface-modified scaffolds on the growth and differentiation of mouse adipose-derived stromal cells. J Tissue Eng Regen Med 2008; 1:211-7. [PMID: 18038413 DOI: 10.1002/term.27] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
PURPOSE Adipose-derived stromal cells (ADSCs) have been shown to increase angiogenesis in ischemic tissue. Maintaining cell survival and facilitating angiogenesis in ischemic tissue, however, continues to be the major challenge of ADSCs implantation. Recently, bioengineered scaffolds were introduced to support and facilitate cell culture and differentiation. The effects of a surface modified three-dimensional (3D) scaffold on ADSC function have not been investigated. Accordingly, the objective of this study was to determine the influence of a gas-plasma treated scaffold on ADSC growth, differentiation into endothelial cell, and angiogenic gene expression. METHODS Freshly isolated mouse ADSCs were characterized by flow cytometry and cultured into wells containing gas-plasma treated scaffolds, non-treated scaffolds, or control wells. Either endothelial growth media or differentiation media was used to alter cell environment. After 3 and 6 days, cell proliferation was analyzed. VEGF concentration in the medium was measured by ELISA. Gene expression was quantified by real-time PCR for VEGF receptor-2 (KDR), cyclooxygenase-2 (COX-2) and matrix metalloproteinases-2 (MMP-2). RESULTS ADSCs expressed stem/endothelial progenitor markers CD34 and CD133 and endothelial cell marker CD31. ADSCs grew in the 3D scaffold. Cells grown on gas-plasma treated scaffolds displayed significantly increased expression of VEGF, COX-2, and MMP-2 when grown in differentiation but not growth media. When cultured in endothelial growth media, VEGF secretion and the expression of KDR, COX-2 and MMP-2 were lower in 3D scaffolds than controls. CONCLUSIONS This study suggests that 3D scaffolds, especially gas-plasma treated scaffolds, support ADSC growth and support differentiation into endothelial cells.
Collapse
Affiliation(s)
- Jing Lin
- Janey Briscoe Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, University of Texas Health Science Center at San Antonio, TX 78229-3900, USA.
| | | | | | | | | |
Collapse
|
22
|
Cai L, Johnstone BH, Cook TG, Liang Z, Traktuev D, Cornetta K, Ingram DA, Rosen ED, March KL. Suppression of hepatocyte growth factor production impairs the ability of adipose-derived stem cells to promote ischemic tissue revascularization. Stem Cells 2007; 25:3234-43. [PMID: 17901400 DOI: 10.1634/stemcells.2007-0388] [Citation(s) in RCA: 159] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The use of adipose-derived stem/stromal cells (ASCs) for promoting repair of tissues is a promising potential therapy, but the mechanisms of their action are not fully understood. We and others previously demonstrated accelerated reperfusion and tissue salvage by ASCs in peripheral ischemia models and have shown that ASCs secrete physiologically relevant levels of hepatocyte growth factor (HGF) and vascular endothelial growth factor. The specific contribution of HGF to ASC potency was determined by silencing HGF expression. RNA interference was used to downregulate HGF expression. A dual-cassette lentiviral construct expressing green fluorescent protein (GFP) and either a small hairpin RNA specifically targeted to HGF mRNA (shHGF) or an inactive control sequence (shCtrl) were used to stably transduce ASCs (ASC-shHGF and ASC-shCtrl, respectively). Transduced ASC-shHGF secreted >80% less HGF, which led to a reduced ability to promote survival, proliferation, and migration of mature and progenitor endothelial cells in vitro. ASC-shHGF were also significantly impaired, compared with ASC-shCtrl, in their ability to promote reperfusion in a mouse hindlimb ischemia model. The diminished ability of ASCs with silenced HGF to promote reperfusion of ischemic tissues was reflected by reduced densities of capillaries in reperfused tissues. In addition, fewer GFP(+) cells were detected at 3 weeks in ischemic limbs of mice treated with ASC-shHGF compared with those treated with ASC-shCtrl. These results indicate that production of HGF is important for the potency of ASCs. This finding directly supports the emerging concept that local factor secretion by donor cells is a key element of cell-based therapies. Disclosure of potential conflicts of interest is found at the end of this article.
Collapse
Affiliation(s)
- Liying Cai
- Indiana Center for Vascular Biology & Medicine, Department of Cellular and Integrative Physiology, Indiana University School of Medicine, 975 W. Walnut Street IB 441, Indianapolis, Indiana 46202, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Mannello F, Tonti GA. Concise Review: No Breakthroughs for Human Mesenchymal and Embryonic Stem Cell Culture: Conditioned Medium, Feeder Layer, or Feeder-Free; Medium with Fetal Calf Serum, Human Serum, or Enriched Plasma; Serum-Free, Serum Replacement Nonconditioned Medium, or Ad Hoc Formula? All That Glitters Is Not Gold! Stem Cells 2007; 25:1603-9. [PMID: 17395775 DOI: 10.1634/stemcells.2007-0127] [Citation(s) in RCA: 233] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The choice of an optimal strategy of stem cell culture is at the moment an impossible task, and the elaboration of a culture medium adapted to the production of embryonic and adult mesenchymal stem cells for the clinical application of cell therapy remains a crucial matter. To make an informed choice, it is crucial to not underestimate the theoretical health risk of using xenogenic compounds, to limit the immunological reactions once stem cells are transplanted, to not overestimate the controversial results obtained with human serum, plasma, and blood derivatives, as well as to carefully examine the pros and cons of serum-free and ad hoc formulation strategies; besides that, to also maintain multipotentiality, self-renewal, and transplantability. The extent to which we are able to achieve effective cell therapies will depend on assimilating a rapidly developing base of scientific knowledge with the practical considerations of design, delivery, and host response. Although clinical studies have already started, many questions remain unsolved, and concomitantly even more evidence on suitable and safe off-the-shelf products (mainly xeno-free) for embryonic and mesenchymal stem cells is cropping up, even though there should be no rush to enter the clinical stage while the underlying basic research is still not so solid; this solely will lead to high-quality translational research, without making blunders stemming from the assumption that all that glitters is not gold. Disclosure of potential conflicts of interest is found at the end of this article.
Collapse
Affiliation(s)
- Ferdinando Mannello
- Institute of Histology and Laboratory Analysis, Faculty of Sciences and Technologies, University of Urbino Carlo Bo, Via O. Ubaldini 7, 61029 Urbino (PU), Italy.
| | | |
Collapse
|
24
|
Ries C, Egea V, Karow M, Kolb H, Jochum M, Neth P. MMP-2, MT1-MMP, and TIMP-2 are essential for the invasive capacity of human mesenchymal stem cells: differential regulation by inflammatory cytokines. Blood 2006; 109:4055-63. [PMID: 17197427 DOI: 10.1182/blood-2006-10-051060] [Citation(s) in RCA: 397] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Human mesenchymal stem cells (hMSCs) represent promising tools in various clinical applications, including the regeneration of injured tissues by endogenous or transplanted hMSCs. The molecular mechanisms, however, that control hMSC mobilization and homing which require invasion through extracellular matrix (ECM) barriers are almost unknown. We have analyzed bone marrow-derivedhMSCs and detected strong expression and synthesis of matrix metalloproteinase 2 (MMP-2), membrane type 1 MMP (MT1-MMP), tissue inhibitor of metalloproteinase 1 (TIMP-1), and TIMP-2. The ability of hMSCs to traverse reconstituted human basement membranes was effectively blocked in the presence of synthetic MMP inhibitors. Detailed studies by RNA interference revealed that gene knock-down of MMP-2, MT1-MMP, or TIMP-2 substantially impaired hMSC invasion, whereas silencing of TIMP-1 enhanced cell migration, indicating opposing roles of both TIMPs in this process. Moreover, the inflammatory cytokines TGF-beta1, IL-1beta, and TNF-alpha up-regulated MMP-2, MT1-MMP, and/or MMP-9 production in these cells, resulting in a strong stimulation of chemotactic migration through ECM, whereas the chemokine SDF-1alpha exhibited minor effects on MMP/TIMP expression and cell invasion. Thus, induction of specific MMP activity in hMSCs by inflammatory cytokines promotes directed cell migration across reconstituted basement membranes in vitro providing a potential mechanism in hMSC recruitment and extravasation into injured tissues in vivo.
Collapse
Affiliation(s)
- Christian Ries
- Division of Clinical Chemistry and Clinical Biochemistry, Surgical Department, Ludwig-Maximillians-University of Munich, Nussbaumstrasse 20, 80336 Munich, Germany.
| | | | | | | | | | | |
Collapse
|