Network


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

Hotspot


Dive into the research topics where Christian Paul is active.

Publication


Featured researches published by Christian Paul.


Stem Cells International | 2015

Exosomes Secreted from CXCR4 Overexpressing Mesenchymal Stem Cells Promote Cardioprotection via Akt Signaling Pathway following Myocardial Infarction

Kai Kang; Ruilian Ma; Wenfeng Cai; Wei Huang; Christian Paul; Jialiang Liang; Yuhua Wang; Tiejun Zhao; Ha Won Kim; Meifeng Xu; Ronald W. Millard; Zhili Wen; Yigang Wang

Background and Objective. Exosomes secreted from mesenchymal stem cells (MSC) have demonstrated cardioprotective effects. This study examined the role of exosomes derived from MSC overexpressing CXCR4 for recovery of cardiac functions after myocardial infarction (MI). Methods. In vitro, exosomes from MSC transduced with lentiviral CXCR4 (ExoCR4) encoding a silencing sequence or null vector were isolated and characterized by transmission electron microscopy and dynamic light scattering. Gene expression was then analyzed by qPCR and Western blotting. Cytoprotective effects on cardiomyocytes were evaluated and effects of exosomes on angiogenesis analyzed. In vivo, an exosome-pretreated MSC-sheet was implanted into a region of scarred myocardium in a rat MI model. Angiogenesis, infarct size, and cardiac functions were then analyzed. Results. In vitro, ExoCR4 significantly upregulated IGF-1α and pAkt levels and downregulated active caspase 3 level in cardiomyocytes. ExoCR4 also enhanced VEGF expression and vessel formation. However, effects of ExoCR4 were abolished by an Akt inhibitor or CXCR4 knockdown. In vivo, ExoCR4 treated MSC-sheet implantation promoted cardiac functional restoration by increasing angiogenesis, reducing infarct size, and improving cardiac remodeling. Conclusions. This study reveals a novel role of exosomes derived from MSCCR4 and highlights a new mechanism of intercellular mediation of stem cells for MI treatment.


Journal of Neuro-oncology | 2003

Postoperative management of patients after stereotactic biopsy: results of a survey of the AANS/CNS Section on Tumors and a single institution study

Ronald E. Warnick; Lynn M. Longmore; Christian Paul; Laurie A. Bode

As little consensus exists on the postoperative care of patients undergoing stereotactic biopsy, we sought to establish a new algorithm for their postoperative management. First, we surveyed active members of the AANS/CNS Section on Tumors to determine national practice patterns for patients after stereotactic biopsy. Second, we retrospectively reviewed 84 consecutive stereotactic biopsy procedures at our institution to assess the potential benefit of routine computed tomography (CT) scanning and intensive care unit (ICU) monitoring. Finally, we prospectively applied this new algorithm in 54 patients to assess its validity. Of 629 surgeons, 263 (42%) responded; they were experienced neurosurgeons (mean 15 years in practice) who performed more than 10 stereotactic biopsies per year. Most surgeons (59%) routinely ordered postoperative CT scans, and the remainder ordered scans based on specific indications. Patients were transferred from the recovery room to a special care unit (47%), regular room (47%), or home (6%). In our retrospective review, 81 patients underwent 84 stereotactic biopsy procedures; 79 underwent postoperative CT scanning and all 81 were monitored overnight in the ICU. Among five (6%) patients who experienced intraoperative hemorrhage, two (2%) underwent craniotomy to control arterial bleeding. Three (4%) patients developed new neurological deficits, which occurred within 2 h of surgery. In both groups, CT scans were helpful in excluding hemorrhage that would require re-operation. In the remaining patients (90%), findings on routine postoperative CT did not alter patient management and ICU monitoring appeared unnecessary because neurological complications occurred within 2 h postoperatively. We confirmed these results in the prospective study of 54 patients undergoing stereotactic biopsy without routine postoperative CT scanning or ICU monitoring. In contrast with national practice patterns reported, we recommend that CT scanning and ICU monitoring be reserved for patients who have intraoperative hemorrhage or new deficits after surgery. All other patients can be monitored for 2 h in the recovery room and transferred to a regular hospital room without a postoperative CT scan.


Oncotarget | 2017

Mesenchymal stem cells release exosomes that transfer miRNAs to endothelial cells and promote angiogenesis

Min Gong; Bin Yu; Jingcai Wang; Yigang Wang; Min Liu; Christian Paul; Ronald W. Millard; De Sheng Xiao; Muhammad Ashraf; Meifeng Xu

Mesenchymal stem cells (MSCs) have been found to benefit patients with a variety of ischemic diseases via promoting angiogenesis. It is also well established that exosomes secreted from MSCs deliver bioactive molecules, including microRNAs (miRs) to recipient cells. Therefore, we hypothesized that exosomes secreted from MSCs deliver miRs into endothelial cells and mediate angiogenesis. The pro-angiogenic stimulatory capacity of exosomes was investigated using tube-like structure formation and spheroid-based sprouting of human umbilical vein endothelial cells (HUVECs), and in vivo Matrigel plug assay. The secretion of pro-angiogenic miRs (pro-angiomiRs) from MSCs into culture medium and transfer of the miRs to HUVECs were confirmed using real-time quantitative PCR. Supplementation of the exosome secretion blocker GW4869 (10 μM) reduced the pro-angiomiRs in the MSC-derived conditioned medium (CdMMSC). Addition of exosomes isolated from CdMMSC could directly 1) promote HUVEC tube-like structure formation in vitro; 2) mobilize endothelial cells into Matrigel plug subcutaneously transplanted into mice; and 3) increase blood flow inside Matrigel plug. Fluorescence tracking showed that the exosomes were internalized rapidly by HUVECs causing an upregulated expression of pro-angiomiRs in HUVECs. Loss-and-gain function of the pro-angiomiRs (e.g., miR-30b) in MSCs significantly altered the pro-angiogenic properties of these MSC-derived exosomes, which could be associated with the regulation of their targets in HUVECs. These results suggest that exosomal transfer of pro-angiogenic miRs plays an important role in MSC mediated angiogenesis and stem cell-to-endothelial cell communication.


Aging Cell | 2016

Blockade of senescence-associated microRNA-195 in aged skeletal muscle cells facilitates reprogramming to produce induced pluripotent stem cells.

Hideyuki Kondo; Ha Won Kim; Lei Wang; Motoi Okada; Christian Paul; Ronald W. Millard; Yigang Wang

The low reprogramming efficiency in cells from elderly patients is a challenge that must be overcome. Recently, it has been reported that senescence‐associated microRNA (miR)‐195 targets Sirtuin 1 (SIRT1) to advance cellular senescence. Thus, we hypothesized that a blockade of miR‐195 expression could improve reprogramming efficiency in old skeletal myoblasts (SkMs). We found that miR‐195 expression was significantly higher in old SkMs (24 months) isolated from C57BL/6 mice as compared to young SkMs (2 months, 2.3‐fold). Expression of SIRT1 and telomerase reverse transcriptase (TERT) was downregulated in old SkMs, and transduction of old SkMs with lentiviral miR‐195 inhibitor significantly restored their expression. Furthermore, quantitative in situ hybridization analysis demonstrated significant telomere elongation in old SkMs transduced with anti‐miR‐195 (1.7‐fold increase). It is important to note that blocking miR‐195 expression markedly increased the reprogramming efficiency of old SkMs as compared to scramble (2.2‐fold increase). Transduction of anti‐miR‐195 did not alter karyotype or pluripotency marker expression. Induced pluripotent stem cells (iPSCs) from old SkMs transduced with anti‐miR‐195 successfully formed embryoid bodies that spontaneously differentiated into three germ layers, indicating that deletion of miR‐195 does not affect pluripotency in transformed SkMs. In conclusion, this study provided novel evidence that the blockade of age‐induced miR‐195 is a promising approach for efficient iPSC generation from aging donor subjects, which has the potential for autologous transplantation of iPSCs in elderly patients.


Stem Cells | 2017

Inhibition of microRNA‐495 Enhances Therapeutic Angiogenesis of Human Induced Pluripotent Stem Cells

Jialiang Liang; Wei Huang; Wenfeng Cai; Lei Wang; Linlin Guo; Christian Paul; Xi-Yong Yu; Yigang Wang

Therapeutic angiogenesis has emerged as a promising strategy to regenerate the damaged blood vessels resulting from ischemic diseases such as myocardial infarction (MI). However, the functional integration of implanted endothelial cells (ECs) in infarcted heart remains challenging. We herein develop an EC generation approach by inhibiting microRNA‐495 (miR‐495) in human induced pluripotent stem cells (hiPSCs) and assess the angiogenic potential for MI treatment. The anti‐angiogenic miR‐495 belonging to Dlk1‐Dio3 miR cluster was identified through expression profiling and computational analysis. Loss‐of‐function experiments for miR‐495 were performed using a lentiviral transfer of antisense sequence in hiPSCs. The pluripotency of hiPSCs was not impacted by the genetic modification. Induced with differentiation medium, miR‐495 inhibition enhanced the expression of EC genes of hiPSCs, as well as the yield of ECs. Newly derived ECs displayed prominent angiogenic characteristics including tube formation, cell migration, and proliferation. Mechanistically, miR‐495 mediated the expression of endothelial or angiogenic genes by directly targeting vascular endothelial zinc finger 1. After transplantation in immunodeficient MI mice, the derived ECs significantly increased neovascularization in the infarcted heart, prevented functional worsening, and attenuated expansion of infarct size. The functional integration of the implanted ECs into coronary networks was also enhanced by inhibiting miR‐495. miR‐495 represents a new target not only for promoting EC generation from hiPSCs but also for enhancing angiogenesis and engraftment of hiPSC‐derived ECs in ischemic heart. Stem Cells 2017;35:337–350


Cell Biochemistry and Function | 2017

Paracrine effect of CXCR4‐overexpressing mesenchymal stem cells on ischemic heart injury

Shizheng Wu; Ying-Lan Li; Wei Huang; Wenfeng Cai; Jialiang Liang; Christian Paul; Lin Jiang; Zhi-Chao Wu; Meifeng Xu; Ping Zhu; Yigang Wang

It has been reported that CXCR4‐overexpressing mesenchymal stem cells (MSCCX4) can repair heart tissue post myocardial infarction. This study aims to investigate the MSCCX4‐derived paracrine cardio‐protective signaling in the presence of myocardial infarction. Mesenchymal stem cells (MSCs) were divided into 3 groups: MSC only, MSCCX4, and CXCR4 gene‐specific siRNA‐transduced MSC. Mesenchymal stem cells were exposed to hypoxia, and then MSCs‐conditioned culture medium was incubated with neonatal and adult cardiomyocytes, respectively. Cell proliferation–regulating genes were assessed by real‐time polymerase chain reaction (RT‐PCR). In vitro: The number of cardiomyocytes undergoing DNA synthesis, cytokinesis, and mitosis was increased to a greater extent in MSCCX4 medium‐treated group than control group, while this proproliferative effect was reduced in CXCR4 gene‐specific siRNA‐transduced MSC–treated cells. Accordingly, the maximal enhancement of vascular endothelial growth factor, cyclin 2, and transforming growth factor‐β2 was observed in hypoxia‐exposed MSCCX4. In vivo: MSCs were labeled with enhanced green fluorescent protein (EGFP) and engrafted into injured myocardium in rats. The number of EGFP and CD31 positive cells in the MSCCX4 group was significantly increased than other 2 groups, associated with the reduced left ventricular (LV) fibrosis, the increased LV free wall thickness, the enhanced angiogenesis, and the improved contractile function. CXCR4 overexpression can mobilize MSCs into ischemic area, whereby these cells can promoted angiogenesis and alleviate LV remodeling via paracrine signaling mechanism.


Journal of Cellular and Molecular Medicine | 2015

CXCR4 attenuates cardiomyocytes mitochondrial dysfunction to resist ischaemia-reperfusion injury

Wenfeng Cai; Kai Kang; Wei Huang; Jialiang Liang; Yuliang Feng; Guan-Sheng Liu; Dehua Chang; Zhili Wen; Christian Paul; Meifeng Xu; Ronald W. Millard; Yigang Wang

The chemokine (C‐X‐C motif) receptor 4 (CXCR4) is expressed on native cardiomyocytes and can modulate isolated cardiomyocyte contractility. This study examines the role of CXCR4 in cardiomyocyte response to ischaemia‐reperfusion (I/R) injury. Isolated adult rat ventricular cardiomyocytes were subjected to hypoxia/reoxygenation (H/R) to simulate I/R injury. In response to H/R injury, the decrease in CXCR4 expression was associated with dysfunctional energy metabolism indicated by an increased adenosine diphosphate/adenosine triphosphate (ADP/ATP) ratio. CXCR4‐overexpressing cardiomyocytes were used to determine whether such overexpression (OE) can prevent bio‐energetic disruption‐associated cell death. CXCR4 OE was performed with adenoviral infection with CXCR4 encoding‐gene or non‐translated nucleotide sequence (Control). The increased CXCR4 expression was observed in cardiomyocytes post CXCR4‐adenovirus transduction and this OE significantly reduced the cardiomyocyte contractility under basal conditions. Although the same extent of H/R‐provoked cytosolic calcium overload was measured, the hydrogen peroxide‐induced decay of mitochondrial membrane potential was suppressed in CXCR4 OE group compared with control group, and the mitochondrial swelling was significantly attenuated in CXCR4 group, implicating that CXCR4 OE prevents permeability transition pore opening exposure to overload calcium. Interestingly, this CXCR4‐induced mitochondrial protective effect is associated with the enhanced signal transducer and activator of transcription 3 (expression in mitochondria. Consequently, in the presence of H/R, mitochondrial dysfunction was mitigated and cardiomyocyte death was decreased to 65% in the CXCR4 OE group as compared with the control group. I/R injury leads to the reduction in CXCR4 in cardiomyocytes associated with the dysfunctional energy metabolism, and CXCR4 OE can alleviate mitochondrial dysfunction to improve cardiomyocyte survival.


PLOS ONE | 2014

Ultrastructural Features of Ischemic Tissue following Application of a Bio-Membrane Based Progenitor Cardiomyocyte Patch for Myocardial Infarction Repair

Dehua Chang; Zhili Wen; Yuhua Wang; Wenfeng Cai; Mashhood Wani; Christian Paul; Teruo Okano; Ronald W. Millard; Yigang Wang

Background and Objective Implantation of cell-sheets into damaged regions of the heart after myocardial infarction (MI) has been shown to improve heart function. However, the tissue morphology following application of induced pluripotent stem cell (iPSC)-derived cardiomyocytes (CM) has not been studied in detail at the level afforded by electron microscopy. We hypothesized that increasing the number of CM derived from iPSC would increase the effectiveness of cell-sheets used to treat ischemic cardiomyopathy. We report here on the ultrastructural features after application of a bio-membrane ‘cell patch’. Methods iPSC-derived progenitor cells were transduced using lentivirus vectors with or without NCX1 promoter. iPSC-CM sheets were transplanted over the transmural MI region in a mouse model of regional ischemic cardiomyopathy. Mice were divided into four groups, 1) Sham; 2) MI; 3) MI + iPSC without NCX1 treated cells (MI + iPSCNull) and 4) MI + iPSC receiving NCX1 promoter treated cells (MI + iPSCNCX1). Echocardiography was performed 4 weeks after cell patch application, followed by histological and transmission electron microscopy (TEM) analysis. Results Large numbers of transplanted CM were observed with significant improvements in left ventricular performance and remodeling in group 4 as compared with group 3. No teratoma formation was detected in any of the treatment groups. Conclusion Manipulation of iPSC yields large numbers of iPSC-CM and favorable morphological and ultrastructural tissue changes. These changes have the potential to enhance current methods used for restoration of cardiac function after MI.


Journal of Molecular and Cellular Cardiology | 2014

Degradation of cardiac myosin light chain kinase by matrix metalloproteinase-2 contributes to myocardial contractile dysfunction during ischemia/reperfusion

Ling Gao; Yan-Jun Zheng; Shanshan Gu; Ji-Liang Tan; Christian Paul; Yigang Wang; Huang-Tian Yang

Although ischemia/reperfusion (I/R)-induced myocardial contractile dysfunction is associated with a prominent decrease in myofilament Ca(2+) sensitivity, the underlying mechanisms have not yet been fully clarified. Phosphorylation of ventricular myosin light chain 2 (MLC-2v) facilitates actin-myosin interactions and enhances contractility, however, its level and regulation by cardiac MLC kinase (cMLCK) and cMLC phosphatase (cMLCP) in I/R hearts are debatable. In this study, the levels and/or effects of MLC-2v phosphorylation, cMLCK, cMLCP, and proteases during I/R were determined. Global myocardial I/R-suppressed cardiac performance in isolated rat hearts was concomitant with decreases of MLC-2v phosphorylation, myofibrillar Ca(2+)-stimulated ATPase activity, and cMLCK content, but not cMLCP proteins. Consistently, simulated I/R in isolated cardiomyocytes inhibited cell shortening, Ca(2+) transients, MLC-2v phosphorylation, and myofilament sensitivity to Ca(2+). These observations were reversed by cMLCK overexpression, while the specific cMLCK knockdown by short hairpin RNA (shRNA) had the opposite effect. Moreover, the inhibition of matrix metalloproteinase-2 (MMP-2, a zinc-dependent endopeptidase) reversed IR-decreased cMLCK, MLC-2v phosphorylation, myofibrillar Ca(2+)-stimulated ATPase activity, myocardial contractile function, and myofilament sensitivity to Ca(2+), while the inhibition or knockdown of cMLCK by ML-9 or specific shRNA abolished MMP-2 inhibition-induced cardioprotection. Finally, the co-localization in cardiomyocytes and interaction in vivo of MMP-2 and cMLCK were observed. Purified recombinant rat cMLCK was concentration- and time-dependently degraded by rat MMP-2 in vitro, and this was prevented by the inhibition of MMP-2. These findings reveal that the I/R-activated MMP-2 leads to the degradation of cMLCK, resulting in a reduction of MLC-2v phosphorylation, and myofibrillar Ca(2+)-stimulated ATPase activity, which subsequently suppresses myocardial contractile function through a decrease of myofilament Ca(2+) sensitivity.


Stem Cells International | 2016

Repair Injured Heart by Regulating Cardiac Regenerative Signals

Wenfeng Cai; Guan-Sheng Liu; Lei Wang; Christian Paul; Zhili Wen; Yigang Wang

Cardiac regeneration is a homeostatic cardiogenic process by which the sections of malfunctioning adult cardiovascular tissues are repaired and renewed employing a combination of both cardiomyogenesis and angiogenesis. Unfortunately, while high-quality regeneration can be performed in amphibians and zebrafish hearts, mammalian hearts do not respond in kind. Indeed, a long-term loss of proliferative capacity in mammalian adult cardiomyocytes in combination with dysregulated induction of tissue fibrosis impairs mammalian endogenous heart regenerative capacity, leading to deleterious cardiac remodeling at the end stage of heart failure. Interestingly, several studies have demonstrated that cardiomyocyte proliferation capacity is retained in mammals very soon after birth, and cardiac regeneration potential is correspondingly preserved in some preadolescent vertebrates after myocardial infarction. There is therefore great interest in uncovering the molecular mechanisms that may allow heart regeneration during adult stages. This review will summarize recent findings on cardiac regenerative regulatory mechanisms, especially with respect to extracellular signals and intracellular pathways that may provide novel therapeutics for heart diseases. Particularly, both in vitro and in vivo experimental evidences will be presented to highlight the functional role of these signaling cascades in regulating cardiomyocyte proliferation, cardiomyocyte growth, and maturation, with special emphasis on their responses to heart tissue injury.

Collaboration


Dive into the Christian Paul's collaboration.

Top Co-Authors

Avatar

Yigang Wang

University of Cincinnati

View shared research outputs
Top Co-Authors

Avatar

Wenfeng Cai

University of Cincinnati

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Meifeng Xu

University of Cincinnati

View shared research outputs
Top Co-Authors

Avatar

Wei Huang

University of Cincinnati

View shared research outputs
Top Co-Authors

Avatar

Jialiang Liang

University of Cincinnati

View shared research outputs
Top Co-Authors

Avatar

Lei Wang

University of Cincinnati

View shared research outputs
Top Co-Authors

Avatar

Zhili Wen

University of Cincinnati

View shared research outputs
Top Co-Authors

Avatar

Dehua Chang

University of Cincinnati

View shared research outputs
Top Co-Authors

Avatar

Guan-Sheng Liu

University of Cincinnati Academic Health Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge