Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
1.
ACS Biomater Sci Eng ; 8(10): 4486-4496, 2022 Oct 10.
Article in English | MEDLINE | ID: mdl-36178141

ABSTRACT

Stromal cell-derived factor-1 alpha (SDF-1α, CXCL12) mediates the migration of circulating cells to desired sites for tissue development, homeostasis, and regeneration and can be used to promote cardiac regeneration by recruiting stem cells. However, the use of SDF-1α in the injured heart necessitates not only higher binding affinity to its receptor, CXCR4+, but also better robustness against enzymatic degradation than other SDF-1 isoforms. Here, we conduct a screening of SDF-1α analog peptides that were designed by structure-based drug design (SBDD), a type of computer-aided drug design (CADD). We have developed in vitro and in vivo methods that enable us to estimate the effect of peptides on the migration of human mesenchymal stem cells (hMSCs) and cardiac regeneration in acute myocardial infarction (AMI)-induced animals, respectively. We demonstrate that one type of SDF-1α analog peptide, SDP-4, among the four analog peptides preselected by SBDD, is more potent than native SDF-1α for cardiac regeneration in myocardial infarction. It is interesting to note that the migratory effects of SDP-4 determined by a wound healing assay, a Transwell assay, and a 2D migration assay are comparable to those of SDF-1α. These results suggest that in vivo, as well as in vitro, screening of peptides developed by SBDD is a quintessential process to the development of a novel therapeutic compound for cardiac regeneration. Our finding also has an implication that the SDP-4 peptide is an excellent candidate for use in the regeneration of an AMI heart.


Subject(s)
Chemokine CXCL12 , Myocardial Infarction , Animals , Cell Movement , Chemokine CXCL12/chemistry , Chemokine CXCL12/pharmacology , Chemokine CXCL12/therapeutic use , Drug Design , Humans , Myocardial Infarction/drug therapy , Myocardial Infarction/metabolism , Peptides/pharmacology , Peptides/therapeutic use , Receptors, CXCR4/metabolism , Receptors, CXCR4/therapeutic use
2.
Biomed J ; 45(3): 522-532, 2022 06.
Article in English | MEDLINE | ID: mdl-35817708

ABSTRACT

BACKGROUND: Stromal cell-derived factor-1α (SDF-1α) is a chemokine associated with tumor progression in various types of cancers. The current study aimed to evaluate whether pre-treatment or kinetics of SDF-1α can predict the prognosis in patients with esophageal squamous cell carcinoma (ESCC) receiving definitive concurrent chemoradiotherapy (CCRT). METHODS: A total of 97 patients with ESCC were identified at Kaohsiung Chang Gung Memorial Hospital between January 2010 and December 2015. Serum concentration of SDF-1α was measured at day 0 (pre-treatment) and chemotherapy day 28 to determine its kinetics and the cut-off level of pre-chemotherapy SDF-1α was 1.5 ng/mL. Two ESCC cell lines, TE1 and KYSE30, were selected to evaluate the function of SDF-1α. RESULTS: Univariate and multivariate analyses showed that pre-treatment SDF-1α ≥ 1.5 ng/mL and an increased SDF-1α level after treatment were significantly associated with worse progression-free survival (p = 0.021 and p = 0.008, respectively) and overall survival (p = 0.005 and p < 0.001, respectively). In addition, patients with pre-treatment SDF-1α ≥ 1.5 ng/mL and increased SDF-1α levels after treatment were found to have poor response to CCRT. Moreover, these cell lines were treated with chemotherapeutic agents (cisplatin or 5-FU) and SDF-1α, alone or in combination. Our in vitro study results showed SDF-1α promoted the proliferation of tumor cells and overcame the cytotoxic effect of chemotherapy (p < 0.001). CONCLUSION: Our study suggested that SDF-1α plays an important role in ESCC disease progression and that pre-treatment SDF-1α level and kinetics of SDF-1α are the independent prognostic factors for ESCC patients receiving definitive CCRT. Periodic determinations of serum SDF-1α level may be valuable to predict prognosis of ESCC in clinical practice.


Subject(s)
Carcinoma, Squamous Cell , Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Carcinoma, Squamous Cell/drug therapy , Chemokine CXCL12/therapeutic use , Chemoradiotherapy/adverse effects , Esophageal Neoplasms/drug therapy , Esophageal Squamous Cell Carcinoma/drug therapy , Esophageal Squamous Cell Carcinoma/etiology , Humans , Prognosis
3.
Neurotherapeutics ; 19(2): 616-634, 2022 03.
Article in English | MEDLINE | ID: mdl-35267171

ABSTRACT

Temozolomide (TMZ) monotherapy is known to be insufficient for resistant/relapsed glioblastoma (GBM), thus seeking a sensitization agent for TMZ is necessary. It was found that regorafenib may improve the overall survival of relapsed GBM patients. We aimed to discover whether regorafenib can enhance the anti-GBM effects of TMZ, and elucidate underlying mechanism. Our analysis of The Cancer Genome Atlas database revealed that the increased expression of CXCR4 is linked to poor survival of GBM patients. Additionally, TMZ treatment may trigger CXCR4/CXCL12 axis of GBM. We used two GBM cell lines, two primary GBM cells, and animal model to identify underlying mechanism and treatment efficacy of regorafenib combined with TMZ by cytotoxicity, apoptosis, reporter gene and invasion/migration assays, chemokine array, Western blotting, MRI, microarray, and immunohistochemistry. We observed that the chemokine CXCL-12 and its receptor CXCR4 regulate the resistance to TMZ, whereas the inhibition of CXCL-12/CXCR4 signaling sensitizes GBM cells to TMZ. The TMZ-induced CXCL-12/CXCR4 signaling, phosphor-extracellular signal-regulated kinases 1 and 2 (ERK1/2) and nuclear factor kappa light chain enhancer of activated B cells (NF-κB), and NF-κB-related proteins can effectively diminish when combining with regorafenib. Regorafenib significantly enhanced the TMZ-induced extrinsic/intrinsic apoptotic pathways, and facilitated the suppression of invasion and migration potential in GBM. Orthotopic tumor experiments demonstrated tumor size reduction and prolonged survival in combination group even with half-dose of TMZ. Our findings provide promising evidence that regorafenib may sensitize GBM to TMZ treatment through inhibition of the CXCL12/CXCR4/ERK/NF-κB signaling.


Subject(s)
Brain Neoplasms , Glioblastoma , Phenylurea Compounds , Pyridines , Animals , Apoptosis , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Line, Tumor , Chemokine CXCL12/pharmacology , Chemokine CXCL12/therapeutic use , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , NF-kappa B/metabolism , Phenylurea Compounds/pharmacology , Phenylurea Compounds/therapeutic use , Pyridines/pharmacology , Pyridines/therapeutic use , Receptors, CXCR4/therapeutic use , Temozolomide/pharmacology , Temozolomide/therapeutic use
4.
Int J Mol Sci ; 22(17)2021 Sep 04.
Article in English | MEDLINE | ID: mdl-34502517

ABSTRACT

Intervertebral disc (IVD) degeneration involves a complex cascade of events, including degradation of the native extracellular matrix, loss of water content, and decreased cell numbers. Cell recruitment strategies for the IVD have been increasingly explored, aiming to recruit either endogenous or transplanted cells. This study evaluates the IVD therapeutic potential of a chemoattractant delivery system (HAPSDF5) that combines a hyaluronan-based thermoreversible hydrogel (HAP) and the chemokine stromal cell derived factor-1 (SDF-1). HAPSDF5 was injected into the IVD and was combined with an intravenous injection of mesenchymal stem/stromal cells (MSCs) in a pre-clinical in vivo IVD lesion model. The local and systemic effects were evaluated two weeks after treatment. The hydrogel by itself (HAP) did not elicit any adverse effect, showing potential to be administrated by intradiscal injection. HAPSDF5 induced higher cell numbers, but no evidence of IVD regeneration was observed. MSCs systemic injection seemed to exert a role in IVD regeneration to some extent through a paracrine effect, but no synergies were observed when HAPSDF5 was combined with MSCs. Overall, this study shows that although the injection of chemoattractant hydrogels and MSC recruitment are feasible approaches for IVD, IVD regeneration using this strategy needs to be further explored before successful clinical translation.


Subject(s)
Chemokine CXCL12/therapeutic use , Hyaluronic Acid/therapeutic use , Intervertebral Disc Degeneration/drug therapy , Administration, Intravenous/methods , Animals , Bone Regeneration/drug effects , Bone Regeneration/physiology , Chemokine CXCL12/administration & dosage , Chemotactic Factors/metabolism , Disease Models, Animal , Extracellular Matrix/metabolism , Hyaluronic Acid/administration & dosage , Hydrogels/therapeutic use , Intervertebral Disc/drug effects , Intervertebral Disc/metabolism , Intervertebral Disc Degeneration/metabolism , Intervertebral Disc Degeneration/physiopathology , Male , Mesenchymal Stem Cells/metabolism , Rats , Rats, Wistar
5.
ACS Appl Mater Interfaces ; 13(31): 36880-36893, 2021 Aug 11.
Article in English | MEDLINE | ID: mdl-34324286

ABSTRACT

Existing local drug delivery systems for periodontitis suffer from poor antibacterial effect and unsatisfied periodontal regeneration. In this study, a smart gingipain-responsive hydrogel (PEGPD@SDF-1) was synthesized as an environmentally sensitive carrier for on-demand drug delivery. The PEGPD@SDF-1 hydrogel was synthesized from polyethylene glycol diacrylate (PEG-DA) based scaffolds, dithiothreitol (DTT), and a novel designed functional peptide module (FPM) via Michael-type addition reaction, and the hydrogel was further loaded with stromal cell derived factor-1 (SDF-1). The FPM exhibiting a structure of anchor peptide-short antimicrobial peptide (SAMP)-anchor peptide could be cleaved by gingipain specifically, and the SAMP was released out of the hydrogel for antibacterial effect in response to gingipain. The hydrogel properties were characterized by scanning electron microscopy (SEM), Fourier transform infrared spectroscopy (FTIR), swelling ratio analysis, degradation evaluation, and release curve description of the SAMP and SDF-1. Results in vitro indicated the PEGPD@SDF-1 hydrogel exhibited preferable biocompatibility and could promote the proliferation, migration, and osteogenic differentiation of periodontal ligament stem cells (PDLSCs). Antibacterial testing demonstrated that the PEGPD@SDF-1 hydrogel released the SAMP stressfully in response to gingipain stimulation, thereby strongly inhibiting the growth of Porphyromonas gingivalis. Furthermore, the study in vivo indicated that the PEGPD@SDF-1 hydrogel inhibited P. gingivalis reproduction, created a low-inflammatory environment, facilitated the recruitment of CD90+/CD34- stromal cells, and induced osteogenesis. Taken together, these results suggest that the gingipain-responsive PEGPD@SDF-1 hydrogel could facilitate in situ periodontal tissue regeneration and is a promising candidate for the on-demand local drug delivery system for periodontitis.


Subject(s)
Bone Regeneration/drug effects , Chemokine CXCL12/therapeutic use , Drug Carriers/chemistry , Gingipain Cysteine Endopeptidases/metabolism , Hydrogels/chemistry , Periodontitis/drug therapy , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/therapeutic use , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/metabolism , Antimicrobial Cationic Peptides/therapeutic use , Cell Differentiation/drug effects , Cell Movement , Chemokine CXCL12/chemistry , Drug Carriers/chemical synthesis , Drug Liberation , Hydrogels/chemical synthesis , Male , Osteogenesis/drug effects , Periodontal Ligament/cytology , Periodontitis/metabolism , Polyethylene Glycols/chemical synthesis , Polyethylene Glycols/chemistry , Polymethacrylic Acids/chemical synthesis , Polymethacrylic Acids/chemistry , Porphyromonas gingivalis/drug effects , Rats, Wistar , Stem Cells
6.
Toxicol Appl Pharmacol ; 427: 115654, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34310909

ABSTRACT

Diabetic cardiomyopathy (DCM) is a serious diabetic complication that lacks effective preventive or therapeutic approaches. Wild-type and Klf15 knockout (Klf15-KO) mice were fed with either high fat diet (HFD, 60% kcal from fat) or normal diet (ND, 10% kcal from fat) for 3 months and then injected with streptozotocin or vehicle, to induce type 2 diabetes (T2D). All T2D and age-matched control mice were treated with or without SDF-1ß at 5 mg/kg body-weight twice a week and also continually received HFD or ND for 3 months. At the end of 6-month study, after cardiac functions were measured, mice were euthanized to collect heart tissue. For in vitro mechanistic study, H9c2 cells were exposed to palmitate to mimic in vivo condition of T2D. SDF-1ß prevented T2D-induced cardiac dysfunction and fibrosis and T2D-down-regulated KLF15 expression in wild-type diabetic heart tissue. However, the preventive effects of SDF-1ß on both KLF15 expression and fibrosis was abolished, with partial cardiac protection in Klf15-KO/T2D mice. These results demonstrate partial KLF15-dependence for SDF-1ß's cardiac fibrotic protection from T2D, but not on SDF-1ß's protective effects on T2D-induced cardiac dysfunction. Further study showed that SDF-1ß inhibited palmitate-induced cardiomyocyte fibrosis through its receptor CXCR7-mediated activation of p38ß MAPK signaling pathway.


Subject(s)
Chemokine CXCL12/therapeutic use , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Diabetic Cardiomyopathies/drug therapy , Kruppel-Like Transcription Factors/deficiency , Animals , Cell Line , Chemokine CXCL12/pharmacology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Diabetic Cardiomyopathies/metabolism , Fibrosis/drug therapy , Fibrosis/metabolism , Kruppel-Like Transcription Factors/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Rats
7.
Eur J Pharm Sci ; 157: 105606, 2021 Feb 01.
Article in English | MEDLINE | ID: mdl-33131745

ABSTRACT

Insufficient T cell infiltration in triple-negative breast cancer (TNBC) has limited its response rate to immune checkpoint blockade (ICB) therapies and motivated the development of immunostimulatory approaches to enhance the ICB therapy. CXCR4 is a chemokine receptor highly upregulated both on cell surface and cytoplasm in tumor tissues. Activating CXCR4 has been associated with increased immunosuppression in the tumor microenvironment. Here, we developed a CXCR4-targeted liposomal formulation (Liposomal-AMD3100) to enhance therapeutic efficacy of AMD3100, a CXCR4 antagonist. Particularly, AMD3100 is not only encapsulated into the liposome but coated on the surface of the formulation to serve as a targeting moiety and a dual blocker capable of inhibiting CXCR4 activation extracellularly and intracellularly. The Liposomal-AMD3100 remodeled both immune and stromal microenvironment more efficiently compared with free AMD3100, indicating better pharmacodynamic profile of AMD3100 achieved by liposomal formulation. The combination of anti-PD-L1 with Liposomal-AMD3100 formulation exhibited an increased antitumor effect and prolonged survival time compared with monotherapies in a murine TNBC model (4T1). This work proves that immune activation via liposomal delivery of CXCR4 inhibitors has a great potential to expand ICB therapies to originally ICB-insensitive cancer types.


Subject(s)
Triple Negative Breast Neoplasms , Animals , Chemokine CXCL12/pharmacology , Chemokine CXCL12/therapeutic use , Humans , Immune Checkpoint Inhibitors , Mice , Receptors, CXCR4/therapeutic use , Signal Transduction , T-Lymphocytes , Triple Negative Breast Neoplasms/drug therapy , Tumor Microenvironment
8.
J Biomed Mater Res A ; 108(8): 1760-1773, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32276293

ABSTRACT

We developed a stromal cell-derived factor-1 alpha (SDF-1α)-aligned silk fibroin (SF)/three-dimensional porous bladder acellular matrix graft (3D-BAMG) composite scaffold for long-section ventral urethral regeneration and repair in vivo. SDF-1α-aligned SF microfiber/3D-BAMG, aligned SF microfiber/3D-BAMG, and nonaligned SF microfiber/3D-BAMG scaffolds were prepared using electrostatic spinning and wet processing. Adipose-derived stem cell (ADSC) and bone marrow stromal cell (BMSC) migration was assessed in the SDF-1α-loaded scaffolds. Sustained SDF-1α release in vitro and vivo was analyzed using enzyme-linked immunosorbent assay (ELISA) and western blotting, respectively. The scaffolds were used to repair a 1.5 × 1 cm2 ventral urethral defect in male rabbits in vivo. General observation and retrograde urinary tract contrast assessment were used to examine urethral lumen patency and continuity at 1 and 3 months post-surgery. Postoperative rehabilitation was evaluated using histological detection. The composite scaffolds sustained SDF-1α release for over 16 days in vitro. SDF-1α-aligned SF nanofiber promoted regeneration of urethral mucosa, submucosal smooth muscles, and microvasculature, increased cellular proliferation, and reduced collagen deposition. SDF-1α expression was increased in reconstructed urethra at 3 months post-surgery in SDF-1α-aligned SF group. SDF-1α-aligned SF microfiber/3D-BAMG scaffolds may be used to repair and reconstruct long urethral defects because they accelerate urethral regeneration.


Subject(s)
Biocompatible Materials/chemistry , Chemokine CXCL12/administration & dosage , Delayed-Action Preparations/chemistry , Fibroins/chemistry , Animals , Cells, Cultured , Chemokine CXCL12/therapeutic use , Male , Rabbits , Plastic Surgery Procedures , Regeneration/drug effects , Tissue Scaffolds/chemistry , Urethra/drug effects , Urethra/physiology , Urethra/surgery
9.
Stem Cell Res Ther ; 10(1): 204, 2019 07 08.
Article in English | MEDLINE | ID: mdl-31286993

ABSTRACT

Endometrial regenerative cells (ERCs) are a new type of mesenchymal-like stromal cells, and their therapeutic potential has been tested in a variety of disease models. SDF-1/CXCR4 axis plays a chemotaxis role in stem/stromal cell migration. The aim of the present study was to investigate the role of SDF-1/CXCR4 axis in the immunomodulation of ERCs on the experimental colitis. The immunomodulation of ERCs in the presence or absence of pretreatment of SDF-1 or AMD3100 was examined in both in vitro cell culture system and dextran sulphate sodium-induced colitis in mice. The results showed that SDF-1 increased the expression of CXCR4 on the surface of ERCs. As compared with normal ERCs, the SDF-1-treated, CXCR4 high-expressing ERCs more significantly suppressed dendritic cell population as well as stimulated both type 2 macrophages and regulatory T cells in vitro and in vivo. Meanwhile, SDF-1-pretreated ERCs increased the generation of anti-inflammatory factors (e.g., IL-4, IL-10) and decreased the pro-inflammatory factors (e.g., IL-6, TNF-α). In addition, SDF-1-pretreated CM-Dil-labeled ERCs were found to engraft to injured colon. Our results may suggest that an SDF-1-induced high level of CXCR4 expression enhances the immunomodulation of ERCs in alleviating experimental colitis in mice.


Subject(s)
Chemokine CXCL12/pharmacology , Colitis/metabolism , Endometrium/cytology , Heterocyclic Compounds/pharmacology , Receptors, CXCR4/metabolism , Animals , Benzylamines , Cells, Cultured , Chemokine CXCL12/therapeutic use , Colitis/chemically induced , Colitis/drug therapy , Cyclams , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Heterocyclic Compounds/therapeutic use , Humans , Male , Mice, Inbred BALB C , Spleen/cytology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism
10.
Neurourol Urodyn ; 37(7): 2141-2150, 2018 09.
Article in English | MEDLINE | ID: mdl-30168617

ABSTRACT

OBJECTIVE: Persistent urinary incontinence (UI) and/or erectile dysfunction (ED) occur in 30-50% of post-radical prostatectomy patients regardless of nerve sparing approaches. Identification of potential treatment options for these patients will require testing in an animal model that develops these chronic conditions. The objective was to characterize a nonhuman primate (NHP) model of persistent post-prostatectomy ED and UI and then test the feasibility of periurethral injection of the chemokine CXCL-12. METHODS: Ten adult male cynomolgus monkeys were used. Two were used for study of normal male nonhuman primate genitourinary anatomy. Five were used for measures of sexual behavior, peak intra-corporal pressure (ICP), abdominal leak point pressures (ALPP) 3 and 6-months post open radical prostatectomy (ORP). Three additional ORP animals received ultrasound-guided peri-urethral injection of chemokine CXCL12 6 weeks after ORP, and UI/ED evaluated for up to 3 months. RESULTS: The anatomy, innervation, and vascular supply to the prostate and surrounding tissues of these male NHPs are substantially similar to those of human beings. ORP resulted in complete removal of the prostate gland along with both neurovascular bundles and seminal vesicles while permitting stable restoration of vesico-urethral patency. ORP produced sustained (6 months) decreases in ALPP, ICP's, and sexual function. Transurethral injection of chemokine CXCL12 was feasible and had beneficial effects on erectile and urinary function. CONCLUSIONS: ORP in NHPs produced persistent erectile and urinary tract dysfunction. Periurethral injection of CXCL-12 was feasible and improved both urinary incontinence and erectile dysfunction and suggests that this model can be used to test new approaches for both conditions.


Subject(s)
Erectile Dysfunction/etiology , Erectile Dysfunction/physiopathology , Postoperative Complications/physiopathology , Prostatectomy/adverse effects , Urinary Incontinence/etiology , Urinary Incontinence/physiopathology , Animals , Chemokine CXCL12/therapeutic use , Disease Models, Animal , Erectile Dysfunction/drug therapy , Feasibility Studies , Macaca fascicularis , Male , Pelvis/anatomy & histology , Postoperative Complications/drug therapy , Sexual Behavior, Animal , Urinary Incontinence/drug therapy , Urodynamics
11.
Cell Mol Neurobiol ; 38(2): 467-477, 2018 Mar.
Article in English | MEDLINE | ID: mdl-28484859

ABSTRACT

Our study aims to investigate the effects of the SDF-1/CXCR4 axis on the repair of traumatic brain injury (TBI) in rats by mediating bone marrow derived from mesenchymal stem cells (BMSCs). Healthy male SD rats were collected, their tibiofibulars were removed, cultured, and BMSCs were collected. The expression of cell-surface molecular proteins was examined using flow cytometry. The mRNA and protein expression of CXCR4 in cells were tested using qRT-PCR and western blotting analysis. An electronic brain injury instrument was utilized to build TBI rat models and each rat was assigned into the experiment, positive control and control groups (10 rats in each group). The morris water maze was used to calculate the escape latency and number of times rats in each group crossed the platform. Neurological severity scores (NSS) was calculated to evaluate the recovery of neurological functioning. The distribution of neuronal nuclear antigens was detected using double-labeling immunohistochemistry. The morphological changes in the hippocampal neuronal and the number of BrdU-positive cells were observed through Nissl's staining and high magnification. The mRNA and protein expressions of CXCR4 were gradually increased as SDF-1 concentration increased. NGF and BDNF positive cells were expressed in each group. The distribution of neuronal nuclear antigens in the experiment group was elevated compared to the control and positive control groups. Among the three groups, the experimental group had the shortest escape latency and the highest number platform crossings. The difference in NSS among the three groups was significant. The experimental group had better cell morphology and a higher number of BrdU-positive cells than the other groups. The present study demonstrates that transplanting BMSCs with SDF-1-induced CXCR4 expression can promote the repair of TBI. This is expected to become a new treatment regimen for TBI.


Subject(s)
Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/therapy , Chemokine CXCL12/therapeutic use , Mesenchymal Stem Cell Transplantation/methods , Receptors, CXCR4/biosynthesis , Animals , Chemokine CXCL12/pharmacology , Dose-Response Relationship, Drug , Male , Maze Learning/drug effects , Maze Learning/physiology , Mesenchymal Stem Cells/metabolism , Rats , Rats, Sprague-Dawley , Treatment Outcome
12.
Cardiovasc Res ; 114(3): 358-367, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29040423

ABSTRACT

Heart failure is rapidly increasing in prevalence and will redraw the global landscape for cardiovascular health. Alleviating and repairing cardiac injury associated with myocardial infarction (MI) is key to improving this burden. Homing signals mobilize and recruit stem cells to the ischaemic myocardium where they exert beneficial paracrine effects. The chemoattractant cytokine SDF-1α and its associated receptor CXCR4 are upregulated after MI and appear to be important in this context. Activation of CXCR4 promotes both cardiomyocyte survival and stem cell migration towards the infarcted myocardium. These effects have beneficial effects on infarct size, and left ventricular remodelling and function. However, the timing of endogenous SDF-1α release and CXCR4 upregulation may not be optimal. Furthermore, current ELISA-based assays cannot distinguish between active SDF-1α, and SDF-1α inactivated by dipeptidyl peptidase 4 (DPP4). Current therapeutic approaches aim to recruit the SDF-1α-CXCR4 pathway or prolong SDF-1α life-time by preventing its cleavage by DPP4. This review assesses the evidence supporting these approaches and proposes SDF-1α as an important confounder in recent studies of DPP4 inhibitors.


Subject(s)
Cardiomyopathies/drug therapy , Chemokine CXCL12/therapeutic use , Heart Failure/drug therapy , Myocardial Ischemia/complications , Myocardium/metabolism , Animals , Cardiomyopathies/etiology , Cardiomyopathies/metabolism , Cardiomyopathies/physiopathology , Chemokine CXCL12/metabolism , Dipeptidyl Peptidase 4/metabolism , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Half-Life , Heart Failure/etiology , Heart Failure/metabolism , Heart Failure/physiopathology , Humans , Myocardial Ischemia/metabolism , Myocardial Ischemia/physiopathology , Myocardium/pathology , Receptors, CXCR4/metabolism , Signal Transduction/drug effects , Ventricular Function, Left/drug effects , Ventricular Remodeling/drug effects
13.
Brain Res ; 1672: 91-105, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28734802

ABSTRACT

Genetic engineering of mesenchymal stem cells (MSCs) and neuronal stem cells (NSCs) has been used to treat spinal cord injuries (SCI). As a mechanism of therapy, MSCs secrete high amounts of trophic factors, while NSCs can differentiate into neuronal lineages and aid in tissue replacement. Additionally, the forced overexpression of secreted proteins can enhance the secretome of transplanted cells, which can increase therapeutic efficacy. This study utilized a combinational treatment consisting of MSCs, NSCs, and the forced overexpression of the chemokine stromal-derived factor-1 (SDF-1) from MSCs (SDF-1-MSCs) as treatment in a rat model of SCI. Transplants occurred at 9-days post-injury, and motor functions were evaluated for 7-weeks post-injury. White matter sparing and axon densities surrounding the lesions were quantified. Findings from this study demonstrate that co-transplanting SDF-1-MSCs with NSCs improved motor functions and enhanced axon densities surrounding the lesion. However, no improvements in white matter sparing were found and tumors were found in some of the animals that received co-transplantations with either SDF-1-MSCs and NSCs or unmodified-MSCs and NSCs, but not in any animal treated with a single cell type. This study offers evidence that providing SDF-1 to NSCs, through the forced expression from MSCs, can enhance the therapeutic potential of the graft, but developing a safe means of doing this requires further work.


Subject(s)
Mesenchymal Stem Cells/cytology , Neural Stem Cells/transplantation , Spinal Cord Injuries/therapy , Animals , Axons/metabolism , Chemokine CXCL12/metabolism , Chemokine CXCL12/therapeutic use , Disease Models, Animal , Female , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/physiology , Neural Stem Cells/metabolism , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Recovery of Function , Spinal Cord/metabolism
14.
Stem Cells Transl Med ; 6(8): 1740-1746, 2017 08.
Article in English | MEDLINE | ID: mdl-28714578

ABSTRACT

Disappointing results of skeletal muscle precursor cell (skMPC) therapy for women with intrinsic urinary sphincter deficiency (ISD) associated urinary incontinence has increased interest in alternative sphincter regenerative approaches. This study was to measure the safety and efficacy of the cell homing chemokine CXCL12 versus skMPCs in a rat model of ISD. Thirty-six adult female Sprague Dawley rats were divided into 6 treatment (Tx) conditions: (a) no ISD/noTx [Control]; (b) ISD/noTx; (c) ISD + skMPCs; (d) ISD + 3.5 mg CXCL12; (e) ISD + 7mg CXCL12; and (f) ISD + 14 mg CXCL12. Tx's were injected directly into the sphincter complex 30 days post ISD and rats euthanized 30 days post Tx. Blood samples for measurements of kidney and liver function, white and red blood cell counts, were taken at baseline and at euthanasia. Leak point pressures (LPP) were measured prior to, and sphincter collagen/muscle content measured after, euthanasia. There were no effects of treatments on white or red/white blood cell counts, kidney/liver function tests or histopathology of the urinary sphincter complex or surrounding tissues. ISD lowered LPP 35% and sphincter muscle content by 17% versus control rats. CXCL12, but not skMPC injections, restored both LPP to control values in a dose-dependent fashion. Both skMPCs and CXCL12 restored sphincter muscle content to control values. This chemokine approach may represent a novel therapeutic option for ISD and appears, at least short-term, to produce little clinical or tissue pathology. Stem Cells Translational Medicine 2017;6:1740-1746.


Subject(s)
Chemokine CXCL12/therapeutic use , Urethral Diseases/drug therapy , Urinary Incontinence/drug therapy , Animals , Blood Cell Count , Chemokine CXCL12/administration & dosage , Chemokine CXCL12/adverse effects , Female , Kidney/physiology , Liver/physiology , Rats , Rats, Sprague-Dawley
15.
J Biomed Mater Res A ; 105(10): 2670-2684, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28571106

ABSTRACT

The design of bioactive scaffolds that can invoke host's own regenerative capabilities and facilitate endogenous tissue repair hold great promise. This study aims to evaluate the potential of stromal cell-derived factor 1 alpha (SDF-1α)-derived peptide and heparin tethered poly(L-lactide-co-ε-caprolactone) (PLCL) copolymers for blood vessel regeneration applications. Amino acid analysis and toluidine blue assays confirm successful conjugation of SDF-1α peptide and heparin with the PLCL copolymers. Assessment of biocompatibility after subcutaneous implantation in rats discloses higher cell infiltration in SDF-1α peptide (SDF-1 group) or SDF-1 peptide and heparin (SDF-1/heparin group) than the control group. SDF-1 and SDF-1/heparin grafts also show more numbers of laminin+ blood vessels, CD90+ stem cells, and alpha smooth muscle actin+ cells than the control group. However, SDF-1 and SDF-1/heparin groups did not significantly differ in terms of blood vessel regeneration and stem cell recruitment. Evaluation of the inflammatory response reveal less numbers of CD68+ macrophages in SDF-1 and SDF-1/heparin groups compared with the control group; whereas three groups show similar numbers of CD206+ macrophages. These results indicate that completely synthetic, cell-free grafts can attract endogenous cells and enhance tissue repair. Bioactive polyesters can be fabricated into different shapes and structures for various tissue engineering applications. © 2017 Wiley Periodicals, Inc. J Biomater Res Part A: 105A: 2670-2684, 2017.


Subject(s)
Biocompatible Materials/therapeutic use , Blood Vessel Prosthesis , Chemokine CXCL12/therapeutic use , Heparin/therapeutic use , Polyesters/therapeutic use , Animals , Biocompatible Materials/chemistry , Blood Vessel Prosthesis Implantation , Chemokine CXCL12/chemistry , Heparin/chemistry , Male , Neovascularization, Physiologic/drug effects , Polyesters/chemistry , Rats, Sprague-Dawley , Tissue Scaffolds/chemistry
16.
Biomaterials ; 134: 13-21, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28453954

ABSTRACT

Traumatic spinal cord injury (SCI) is a devastating event for which functional recovery remains elusive. Due to the complex nature of SCI pathology, a combination treatment strategy will likely be required for success. We hypothesized that tissue and functional repair would be achieved in a rat model of impact-compression SCI by combining degradation of the glial scar, using chondroitinase ABC (ChABC), with recruitment of endogenous neural precursor cells (NPCs), using stromal cell-derived factor 1α (SDF). To test this hypothesis, we designed a crosslinked methylcellulose hydrogel (XMC) for minimally invasive, localized, and sustained intrathecal drug delivery. ChABC was released from XMC using protein-peptide affinity interactions while SDF was delivered by electrostatic affinity interactions from polymeric nanoparticles embedded in XMC. Rats with SCI were treated acutely with a combination of SDF and ChABC, SDF alone, ChABC alone, or vehicle alone, and compared to injury only. Treatment with ChABC, both alone and in combination with SDF, resulted in faster and more sustained behavioural improvement over time than other groups. The significantly reduced chondroitin sulfate proteoglycan levels and greater distribution of NPCs throughout the spinal cord tissue with ChABC delivery, both alone and in combination with SDF, may explain the improved locomotor function. Treatment with SDF alone had no apparent effect on NPC number or distribution nor synergistic effect with ChABC delivery. Thus, in this model of SCI, tissue and functional repair is attributed to ChABC.


Subject(s)
Chemokine CXCL12/chemistry , Chondroitin ABC Lyase/metabolism , Spinal Cord Injuries/metabolism , Animals , Chemokine CXCL12/metabolism , Chemokine CXCL12/therapeutic use , Chondroitin ABC Lyase/chemistry , Chondroitin Sulfate Proteoglycans/chemistry , Enzyme-Linked Immunosorbent Assay , Female , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Immunohistochemistry , Methylcellulose/chemistry , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Rats , Rats, Sprague-Dawley , Spinal Cord Injuries/drug therapy
18.
Neuroimmunomodulation ; 24(6): 331-340, 2017.
Article in English | MEDLINE | ID: mdl-29680839

ABSTRACT

OBJECTIVE: The chemokine CXCL12 interacting with the CXC receptor 4 (CXCR4) has been reported to play a role in the development and progression of bronchial asthma, but its mechanism of action is still unknown. The objective of this study was to assess the effect of the CXCL12 neutraligand chalcone 4 on the migration of dendritic cells (DCs) in a murine model of allergic airway inflammation. METHODS: A 21-day ovalbumin (OVA)-induced allergic-airway TH2 inflammation model in BALB/c mice was used. Four groups were sensitized with OVA adsorbed on alum and challenged either with OVA or saline for 4 days. Mice were treated intranasally with chalcone 4 (300 nmol/kg body weight) or solvent 2 h before each OVA or saline challenge; 24 h after the last challenge, CD11c+F4/80- DCs were counted in the bronchoalveolar lavage. Jugular-nodose ganglion complex (JNC) sections were sampled, and for immunofluorescence staining, cryocut sections were prepared. MHC II+F4/80- DCs as well as calcitonin gene-related peptide (CGRP)- and substance P (SP)-positive neuronal cell bodies were analyzed. RESULTS: In OVA-challenged mice, chalcone 4 caused a significantly decreased DC/neuron ratio in the JNC from 51.7% in solvent-treated to 32.6% in chalcone 4-treated mice. In parallel, chalcone 4 also decreased the DC population in BALF from 11.5 × 103 cells in solvent to 4.5 × 103 cells in chalcone 4-treated mice. By contrast, chalcone 4 had no effect on the expression of the neuropeptides CGRP and SP in JNC. CONCLUSION: This study reported the CXCL12 neutraligand chalcone 4 to affect DC infiltration into the airways and airway ganglia as well as to decrease airway eosinophilic inflammation and, therefore, validated CXCL12 as a new target in allergic disease models of asthma.


Subject(s)
Asthma/immunology , Cell Movement/immunology , Chalcone/pharmacology , Chemokine CXCL12/pharmacology , Dendritic Cells/immunology , Nodose Ganglion/immunology , Animals , Asthma/chemically induced , Asthma/drug therapy , Cell Movement/drug effects , Chalcone/therapeutic use , Chemokine CXCL12/therapeutic use , Dendritic Cells/drug effects , Ligands , Male , Mice , Mice, Inbred BALB C , Nodose Ganglion/cytology , Nodose Ganglion/drug effects , Ovalbumin/toxicity
19.
J Control Release ; 238: 114-122, 2016 09 28.
Article in English | MEDLINE | ID: mdl-27473766

ABSTRACT

Diabetic foot ulcers (DFUs) are a severe complication of diabetes mellitus. Altered cell migration due to microcirculatory deficiencies as well as excessive and prolonged reactive oxygen species production are implicated in the delayed healing of DFUs. The goal of this research was to assess whether sustained release of SDF-1, a chemokine that promotes endothelial progenitor cell homing and angiogenesis, from a citrate-based antioxidant thermoresponsive polymer would significantly improve impaired dermal wound healing in diabetes. Poly (polyethylene glycol citrate-co-N-isopropylacrylamide) (PPCN) was synthesized via sequential polycondensation and free radical polymerization reactions. SDF-1 was entrapped via gelation of the PPCN+SDF-1 solution above its lower critical solution temperature (LCST) and its release and bioactivity was measured. The effect of sustained release of SDF-1 from PPCN (PPCN+SDF-1) versus a bolus application of SDF-1 in phosphate buffered saline (PBS) on wound healing was evaluated in a diabetic murine splinted excisional dermal wound model using gross observation, histology, immunohistochemistry, and optical coherence tomography microangiography. Increasing PPCN concentration decreased SDF-1 release rate. The time to 50% wound closure was 11days, 16days, 14days, and 17days for wounds treated with PPCN+SDF-1, SDF-1 only, PPCN only, and PBS, respectively. Wounds treated with PPCN+SDF-1 had the shortest time for complete healing (24days) and exhibited accelerated granulation tissue production, epithelial maturation, and the highest density of perfused blood vessels. In conclusion, sustained release of SDF-1 from PPCN is a promising and easy to use therapeutic strategy to improve the treatment of chronic non-healing DFUs.


Subject(s)
Antioxidants/chemistry , Chemokine CXCL12/administration & dosage , Citrates/chemistry , Delayed-Action Preparations/chemistry , Diabetic Foot/drug therapy , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Wound Healing/drug effects , Animals , Cell Line , Cell Movement/drug effects , Chemokine CXCL12/therapeutic use , Diabetic Foot/pathology , Humans , Temperature
20.
J Urol ; 196(6): 1809-1815, 2016 12.
Article in English | MEDLINE | ID: mdl-27267321

ABSTRACT

PURPOSE: Mixed efficacy results of autologous skeletal muscle precursor cell therapy in women with chronic intrinsic urinary sphincter deficiency have increased interest in the therapeutic value of alternative regenerative medicine approaches. The goal of this study was to compare the effects of the cell homing chemokine CXCL12 (C-X-C motif chemokine 12) and skeletal muscle precursor cells on chronic urinary sphincter regeneration in chronic intrinsic urinary sphincter deficiency. MATERIALS AND METHODS: Five million autologous skeletal muscle precursor cells or 100 ng CXCL12 were injected in the urinary sphincter complex of adult female cynomolgus monkeys with chronic (6-month history) intrinsic urinary sphincter deficiency. These treatment groups of 3 monkeys per group were compared to a group of 3 with no intrinsic urinary sphincter deficiency and no injection, and a group of 3 with intrinsic urinary sphincter deficiency plus vehicle injection. Maximal urethral pressure was measured at rest, during stimulation of the urinary sphincter pudendal nerves at baseline and again 6 months after treatment. The monkeys were then necropsied. The urinary sphincters were collected for tissue analysis of muscle and collagen content, vascularization and motor endplates. RESULTS: CXCL12 but not skeletal muscle precursor cells increased resting maximal urethral pressure in nonhuman primates with chronic intrinsic urinary sphincter deficiency compared to that in monkeys with intrinsic urinary sphincter plus vehicle injection (p >0.05). Skeletal muscle precursor cells and CXCL12 only partially restored pudendal nerve stimulated increases in maximal urethral pressure (p >0.05), sphincter vascularization and motor endplate expression in monkeys with chronic intrinsic urinary sphincter deficiency. Additionally, CXCL12 but not skeletal muscle precursor cell injections decreased collagen and increased the muscle content of urinary sphincter complex in monkeys with chronic intrinsic urinary sphincter deficiency compared to those with intrinsic urinary sphincter plus vehicle injection and no intrinsic urinary sphincter plus no injection (p <0.05 and >0.05, respectively). CONCLUSIONS: These results raise questions about cell therapy for chronic intrinsic urinary sphincter deficiency and identify a chemokine treatment (CXCL12) as a potential alternative treatment of chronic intrinsic urinary sphincter deficiency.


Subject(s)
Chemokine CXCL12/therapeutic use , Myoblasts/transplantation , Urethral Diseases/drug therapy , Urethral Diseases/surgery , Animals , Chronic Disease , Disease Models, Animal , Female , Macaca fascicularis
SELECTION OF CITATIONS
SEARCH DETAIL
...