Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Fa-Ming Chen is active.

Publication


Featured researches published by Fa-Ming Chen.


Stem Cell Research & Therapy | 2016

Treatment of periodontal intrabony defects using autologous periodontal ligament stem cells: a randomized clinical trial

Fa-Ming Chen; Li-Na Gao; Bei-Min Tian; Xi-Yu Zhang; Yongjie Zhang; Guangying Dong; Hong Lu; Qing Chu; Jie Xu; Yang Yu; Rui-Xin Wu; Yuan Yin; Songtao Shi; Yan Jin

BackgroundPeriodontitis, which progressively destroys tooth-supporting structures, is one of the most widespread infectious diseases and the leading cause of tooth loss in adults. Evidence from preclinical trials and small-scale pilot clinical studies indicates that stem cells derived from periodontal ligament tissues are a promising therapy for the regeneration of lost/damaged periodontal tissue. This study assessed the safety and feasibility of using autologous periodontal ligament stem cells (PDLSCs) as an adjuvant to grafting materials in guided tissue regeneration (GTR) to treat periodontal intrabony defects. Our data provide primary clinical evidence for the efficacy of cell transplantation in regenerative dentistry.MethodsWe conducted a single-center, randomized trial that used autologous PDLSCs in combination with bovine-derived bone mineral materials to treat periodontal intrabony defects. Enrolled patients were randomly assigned to either the Cell group (treatment with GTR and PDLSC sheets in combination with Bio-oss®) or the Control group (treatment with GTR and Bio-oss® without stem cells). During a 12-month follow-up study, we evaluated the frequency and extent of adverse events. For the assessment of treatment efficacy, the primary outcome was based on the magnitude of alveolar bone regeneration following the surgical procedure.ResultsA total of 30 periodontitis patients aged 18 to 65 years (48 testing teeth with periodontal intrabony defects) who satisfied our inclusion and exclusion criteria were enrolled in the study and randomly assigned to the Cell group or the Control group. A total of 21 teeth were treated in the Control group and 20 teeth were treated in the Cell group. All patients received surgery and a clinical evaluation. No clinical safety problems that could be attributed to the investigational PDLSCs were identified. Each group showed a significant increase in the alveolar bone height (decrease in the bone-defect depth) over time (pu2009<u20090.001). However, no statistically significant differences were detected between the Cell group and the Control group (pu2009>u20090.05).ConclusionsThis study demonstrates that using autologous PDLSCs to treat periodontal intrabony defects is safe and does not produce significant adverse effects. The efficacy of cell-based periodontal therapy requires further validation by multicenter, randomized controlled studies with an increased sample size.Trial RegistrationNCT01357785 Date registered: 18 May 2011.


Journal of Materials Chemistry B | 2016

Directing immunomodulation using biomaterials for endogenous regeneration

Yang Yu; Rui-Xin Wu; Yuan Yin; Fa-Ming Chen

Stem cell therapy and tissue engineering hold considerable potential for innovative and transformative strategies to repair damaged tissue form and function. Although many approaches are adopting ex vivo expanded cells for transplantation, an alternative is to manipulate the biomaterial-host interactions that recruit the patients own stem cells endogenously for regeneration. There are several considerations in targeting the biomaterial-host interactions therapeutically, not the least of which is the biomimetic design of extracellular matrix (ECM)-mimicking materials and the administration of navigation cues and small molecules that target specific aspects of the native healing cascades to stimulate homing of endogenous stem cells and, thereafter, their expansion and differentiation. A sequence of coordinated interactions between the local niche cells and implanted biomaterials offers signals and sign posts that may instruct the cells traveling toward the injured tissues. Furthermore, stem cell function is critically influenced by extrinsic signals provided by the niche as well as by the implanted biomaterials. Novel strategies harnessing growth factors and immunological cues to design materials not only can modulate the behavior of stem cells but also can alter innate and adaptive immunity in a controlled manner. We envisage that successful and safe endogenous regeneration will involve at least three aspects, i.e., homing of sufficient stem cells, controlling cell fate determination, and blunting host immune responses to outside biomaterial devices. Improving our understanding of the biological and physicochemical signals of biomimetic biomaterials that govern immunomodulation for in situ tissue regeneration, particularly context-dependent macrophage (Mφ) polarization, will lead to a concurrent improvement in clinical outcomes.


Advanced Biosystems | 2017

Engineering a Cell Home for Stem Cell Homing and Accommodation

Rui-Xin Wu; Yuan Yin; Xiao-Tao He; Xuan Li; Fa-Ming Chen

Distilling complexity to advance regenerative medicine from laboratory animals to humans, in situ regeneration will continue to evolve using biomaterial strategies to drive endogenous cells within the human body for therapeutic purposes; this approach avoids the need for delivering ex vivo‐expanded cellular materials. Ensuring the recruitment of a significant number of reparative cells from an endogenous source to the site of interest is the first step toward achieving success. Subsequently, making the “cell home” cell‐friendly by recapitulating the natural extracellular matrix (ECM) in terms of its chemistry, structure, dynamics, and function, and targeting specific aspects of the native stem cell niche (e.g., cell–ECM and cell–cell interactions) to program and steer the fates of those recruited stem cells play equally crucial roles in yielding a therapeutically regenerative solution. This review addresses the key aspects of material‐guided cell homing and the engineering of novel biomaterials with desirable ECM composition, surface topography, biochemistry, and mechanical properties that can present both biochemical and physical cues required for in situ tissue regeneration. This growing body of knowledge will likely become a design basis for the development of regenerative biomaterials for, but not limited to, future in situ tissue engineering and regeneration.


Journal of Tissue Engineering and Regenerative Medicine | 2017

Platelet lysate supports the in vitro expansion of human periodontal ligament stem cells for cytotherapeutic use

Rui-Xin Wu; Yang Yu; Yuan Yin; Xi-Yu Zhang; Li-Na Gao; Fa-Ming Chen

Human platelet lysate (PL) produced under optimal conditions of standardization and safety has been increasingly suggested as the future ‘gold standard’ supplement to replace fetal bovine serum (FBS) for the ex vivo propagation of mesenchymal stem cells for translational medicine and cell therapy applications. However, the multifaceted effects of PL on tissue‐specific stem cells remain largely unexplored. In the present study, we investigated the stem cell behaviours of human periodontal ligament stem cells (PDLSCs) in media with or without PL. Our data indicate that human PL, either as an adjuvant for culture media or as a substitute for FBS, supports the proliferation and expansion of human PDLSCs derived from either ‘young’ or ‘old’ donors to the same extent as FBS, without interfering with their immunomodulatory capacities. Although PL appears to inhibit the in vitro differentiation of ‘young’ or ‘old’ PDLSCs, their decreased osteogenic potential may be restored to similar or higher levels compared with FBS‐expanded cells. PL‐ and FBS‐expanded PDLSCs exhibited a similar potential to form mineralized nodules and expressed similar levels of osteogenic genes. Our data indicate that large clinically relevant quantities of PDLSCs may be yielded by the use of human PL; however, further analysis of its precise composition and function will pave the way for determining optimized, defined culture conditions. In addition to the potential increase in patient safety, our findings highlight the need for further research to develop the potential of PL‐expanded PDLSCs for clinical use. Copyright


Stem Cell Research & Therapy | 2017

Influences of age-related changes in mesenchymal stem cells on macrophages during in-vitro culture

Yuan Yin; Rui-Xin Wu; Xiao-Tao He; Xin-Yue Xu; Jia Wang; Fa-Ming Chen

BackgroundMesenchymal stem cells (MSCs) have been widely used in cytotherapy and tissue engineering due to their immunosuppressive ability and regenerative potential. Recently, the immunomodulatory influence of MSCs has been gaining increasing attention because their functional roles in modulating immune responses likely have high clinical significance.MethodsIn this study, we investigated the influence of MSCs on macrophages (Mφs) in in-vitro cell culture systems. Given evidence that aged MSCs are functionally compromised, bone marrow-derived MSCs (BMSCs) isolated from both young and aged mice (YMSCs and AMSCs) were evaluated and contrasted.ResultsWe found that YMSCs exhibited greater proliferative and osteo-differentiation potential compared to AMSCs. When cocultured with RAW264.7 cells (an Mφ cell line), both YMSCs and AMSCs coaxed polarization of Mφs toward an M2 phenotype and induced secretion of anti-inflammatory and immunomodulatory cytokines. Compared to AMSCs, YMSCs exhibited a more potent immunomodulatory effect. While Mφs cocultured with either YMSCs or AMSCs displayed similar phagocytic ability, AMSC coculture was found to enhance Mφ migration in Transwell systems. When BMSCs were prestimulated with interferon gamma before coculture with RAW264.7 cells, their regulatory effects on Mφs appeared to be modified. Here, compared to stimulated AMSCs, stimulated YMSCs also exhibited enhanced cellular influence on cocultured RAW264.7 cells.ConclusionsOur data suggest that BMSCs exert an age-related regulatory effect on Mφs with respect to their phenotype and functions but an optimized stimulation to enhance MSC immunomodulation is in need of further investigation.


Stem Cell Research & Therapy | 2017

Exosomes secreted by stem cells from human exfoliated deciduous teeth contribute to functional recovery after traumatic brain injury by shifting microglia M1/M2 polarization in rats

Ye Li; Yuan-Yuan Yang; Jiali Ren; Feng Xu; Fa-Ming Chen; Ang Li

BackgroundTraumatic brain injury (TBI) is one of the major causes of mortality and disability for all ages worldwide. Mesenchymal stem cells (MSCs)-originated exosomes have provided therapeutic effects. However, as an indispensable component of MSCs, whether odontogenic stem cell-generated exosomes could benefit TBI is still unclear. Thus we aimed to explore the potential of stem cells from human exfoliated deciduous teeth-originated exosomes (SHED-Ex) for the management of TBI.MethodsFirst, a transwell system was used to co-culture activated BV-2 microglia cells with SHED. The secretion levels of neuroinflammatory factors and nitrite were evaluated by enzyme-linked immunosorbent assay (ELISA) and Griess assay. Furthermore, purified SHED-Ex were co-cultured with activated BV-2. ELISA, Griess assay, flow cytometry, immunofluorescence, and qRT-PCR were performed to test the levels of inflammatory factors as well as the microglia phenotype. Finally, SHED and SHED-Ex were locally injected into TBI rat models. Basso, Beattie, and Bresnahan (BBB) scores were chosen to evaluate the motor functional recovery. Histopathology and immunofluorescence were performed to measure the lesion volume and neuroinflammation.ResultsAs a result, SHED-Ex could reduce neuroinflammation by shifting microglia polarization. The administration of SHED-Ex improves rat motor functional recovery and reduces cortical lesion compared with the control group 2xa0weeks post-injury (Pu2009<u20090.05).ConclusionsThe current study demonstrates for the first time that SHED-Ex contribute a therapeutic benefit to TBI in rats, at least in part by shifting microglia polarization to reduce neuroinflammation. The use of odontogenic stem cells, and indeed their exosomes, may be expanded for the treatment of TBI or other neurological disorders.


Journal of Dental Research | 2017

Leveraging Stem Cell Homing for Therapeutic Regeneration

Yuan Yin; Xuan Li; Xiao-Tao He; Rui-Xin Wu; Hai-Hua Sun; Fa-Ming Chen

Resident stem cell pools in many tissues/organs are responsible not only for tissue maintenance during physiologic turnover but also for the process of wound repair following injury. With inspiration from stem cell trafficking within the body under physiologic and pathologic conditions, recent advances have been made toward inducing stem cell mobilization and directing patients’ own cells to sites of interest for treating a broad spectrum of diseases. An evolving body of work corroborates that delivering guidance cues can mobilize stem cells from the bone marrow and drive these cells toward a specific region. In addition, the transplantation of cell-friendly biomaterials incorporating certain biomolecules has led to the regeneration of lost/damaged tissue without the need for delivering cellular materials manipulated ex vivo. Recently, cell homing has resulted in remarkable biological discoveries in the laboratory as well as great curative successes in preclinical scenarios. Here, we review the biological evidence underlying in vivo cell mobilization and homing with the aim of leveraging endogenous reparative cells for therapeutic applications. Considering both the promise and the obstacles of this approach, we discuss how matrix components of the in vivo milieu can be modified to promote the native regenerative process and inspire future tissue-engineering design.


Cell and Tissue Research | 2016

Effects of short-term inflammatory and/or hypoxic pretreatments on periodontal ligament stem cells: in vitro and in vivo studies

Yang Yu; Chun-Sheng Bi; Rui-Xin Wu; Yuan Yin; Xi-Yu Zhang; Ping-Heng Lan; Fa-Ming Chen

In this study, we extensively screened the in vitro and in vivo effects of PDLSCs following short-term inflammatory and/or hypoxic pretreatments. We found that the 24-h hypoxic pretreatment of PDLSCs significantly enhanced cell migration and improved cell surface CXCR4 expression. In addition, hypoxia-pretreated PDLSCs exhibited improved cell colony formation and proliferation. Cells that were dually stimulated also formed more colonies compared to untreated cells but their proliferation did not increase. Importantly, the hypoxic pretreatment of PDLSCs enhanced cell differentiation as determined by elevated RUNX-2 and ALP protein expression. In this context, the inflammatory stimulus impaired cell OCN protein expression, while dual stimuli led to decreased RUNX-2 and OCN mRNA levels. Although preconditioning PDLSCs with inflammatory and/or hypoxic pretreatments resulted in no differences in the production of matrix proteins, hypoxic pretreatment led to the generation of thicker cell sheets; the inflammatory stimulus weakened the ability of cells to form sheets. All the resultant cell sheets exhibited clear bone regeneration following ectopic transplantation as well as in periodontal defect models; the amount of new bone formed by hypoxia-preconditioned cells was significantly greater than that formed by inflammatory stimulus- or dual-stimuli-treated cells or by nonpreconditioned cells. The regeneration of new cementum and periodontal ligaments was only identified in the hypoxia-stimulus and no-stimulus cell groups. Our findings suggest that PDLSCs that undergo short-term hypoxic pretreatment show improved cellular behavior in vitro and enhanced regenerative potential in vivo. The preconditioning of PDLSCs via combined treatments or an inflammatory stimulus requires further investigation.


Cell Proliferation | 2017

Hypoxia and low‐dose inflammatory stimulus synergistically enhance bone marrow mesenchymal stem cell migration

Yang Yu; Yuan Yin; Rui-Xin Wu; Xiao-Tao He; Xi-Yu Zhang; Fa-Ming Chen

Cell migration is necessary for numerous physiological cell processes. Although either inflammatory or hypoxic stimuli of certain dose and duration have positive influence on cell migration, their combination has not been shown to result in a synergistic effect.


Acta Biomaterialia | 2018

Macrophage involvement affects matrix stiffness-related influences on cell osteogenesis under three-dimensional culture conditions

Xiao-Tao He; Rui-Xin Wu; Xin-Yue Xu; Jia Wang; Yuan Yin; Fa-Ming Chen

Accumulating evidence indicates that the physicochemical properties of biomaterials exert profound influences on stem cell fate decisions. However, matrix-based regulation selected through in vitro analyses based on a given cell population do not genuinely reflect the in vivo conditions, in which multiple cell types are involved and interact dynamically. This study constitutes the first investigation of how macrophages (Mφs) in stiffness-tunable transglutaminase cross-linked gelatin (TG-gel) affect the osteogenesis of bone marrow-derived mesenchymal stem cells (BMMSCs). When a single cell type was cultured, low-stiffness TG-gels promoted BMMSC proliferation, whereas high-stiffness TG-gels supported cell osteogenic differentiation. However, Mφs in high-stiffness TG-gels were more likely to polarize toward the pro-inflammatory M1 phenotype. Using either conditioned medium (CM)-based incubation or Transwell-based co-culture, we found that Mφs encapsulated in the low-stiffness matrix exerted a positive effect on the osteogenesis of co-cultured BMMSCs. Conversely, Mφs in high-stiffness TG-gels negatively affected cell osteogenic differentiation. When both cell types were cultured in the same TG-gel type and placed into the Transwell system, the stiffness-related influences of Mφs on BMMSCs were significantly altered; both the low- and high-stiffness matrix induced similar levels of BMMSC osteogenesis. Although the best material parameter for synergistically affecting Mφs and BMMSCs remains unknown, our data suggest that Mφ involvement in the co-culture system alters previously identified material-related influences on BMMSCs, such as matrix stiffness-related effects, which were identified based on a culture system involving a single cell type. Such Mφ-stem cell interactions should be considered when establishing proper matrix parameter-associated cell regulation in the development of biomimetic biomaterials for regenerative applications.nnnSTATEMENT OF SIGNIFICANCEnThe substrate stiffness of a scaffold plays critical roles in modulating both reparative cells, such as mesenchymal stem cells (MSCs), and immune cells, such as macrophages (Mφs). Although the influences of material stiffness on either Mφs or MSCs, have been extensively described, how the two cell types respond to matrix cues to dynamically affect each other in a three-dimensional (3D) biosystem remains largely unknown. Here, we report our findings that, in a platform wherein Mφs and bone marrow-derived MSCs coexist, matrix stiffness can influence stem cell fate through both direct matrix-associated regulation and indirect Mφ-based modulation. Our data support future studies of the MSC-Mφ-matrix interplay in the 3D context to optimize matrix parameters for the development of the next biomaterial.

Collaboration


Dive into the Fa-Ming Chen's collaboration.

Top Co-Authors

Avatar

Rui-Xin Wu

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Yuan Yin

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Xiao-Tao He

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Bei-Min Tian

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Yang Yu

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Xuan Li

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Xi-Yu Zhang

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Xin-Yue Xu

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Jia Wang

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Li-Na Gao

Fourth Military Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge