Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
Biophys J ; 123(7): 782-798, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38341756

ABSTRACT

A spiral wavefront (WF), generated by a cardiac rotor that drifts between surface electrodes during atrial fibrillation, exhibits frequency changes inconsistent with classical Doppler effect (CDE) phenomena. Recent clinical studies reveal three repeatedly observed events--1) side-dependent frequency changes across the path of the rotor, 2) one additional WF strike on the higher frequency side, and 3) a reversal of WF strike sequence--which constitute a diametrical property of spinning WF sources. A linear ray model is first used to reveal and develop the diametrical phenomena. Mathematical models of an Archimedean spiral and a spiral generated by the diffusion equation are developed and compared. Each formulation predicts the diametrical property that CDE does not capture and illuminates the occurrence of a strong side and weak side with respect to the rotor path. Whereas CDE exhibits higher and lower frequencies from approaching and receding sources of WFs, respectively, spiral rotors generate higher and lower frequencies on opposite sides of the migration path. This motivates the reconsideration of mapping and ablation strategies that have traditionally been based on identifying sites of the dominant frequency. While this research aims to characterize the path of a spiral rotor during atrial fibrillation accurately, the results are applicable in other fields of science and engineering in which rotating spiral waves occur.


Subject(s)
Atrial Fibrillation , Humans , Atrial Fibrillation/surgery , Heart Conduction System , Models, Theoretical , Heart , Diffusion
2.
Front Reprod Health ; 5: 1244659, 2023.
Article in English | MEDLINE | ID: mdl-37744288

ABSTRACT

There is strong global need for the development of Multipurpose Prevention Technologies (MPTs) that prevent HIV, pregnancy, and/or other sexually transmitted infections (STIs). However, despite decades of research focused on the development of MPTs, numerous research gaps remain, contributing to reproductive health disparities. This commentary will highlight biomedical, socio-behavioral, and implementation science gaps in MPT research. Biomedical gaps and barriers include limited dosage forms, challenges around drug selection and stable coformulation of multiple drugs, and an unclear regulatory pathway. Behavioral, social, and structural gaps include lack of research around MPT preferences for some subgroups of potential end users, lack of knowledge around whether MPTs improve uptake, adherence, and persistence vs. separate products, and a need to further understand how social and cultural factors might impact MPT interest and use. Gaps in implementation science research will need to be addressed to better understand how to implement MPTs to maximize effectiveness and benefit. This commentary will also identify opportunities for integrating biomedical and behavioral science around MPTs.

3.
J Environ Manage ; 338: 117742, 2023 Jul 15.
Article in English | MEDLINE | ID: mdl-37001426

ABSTRACT

Since 2012, control of invasive wild pigs (Sus scrofa) in the United States (US) has become a state and national priority due to their propensity to damage agricultural commodities and infrastructure, transmit disease, detrimentally affect ecological processes, and compete with native wildlife for resources. While several life-history characteristics certainly aided their proliferation, the recreational value of wild pigs was likely the stimulus for translocation and subsequent establishment of populations in ≥35 states, causing an annual economic burden of $1.5 billion in the US. Consequently, state-level legislative procedures regarding wild pigs are expanding in scope and priority, but policy among states lacks uniformity. States vary in their treatment of wild pig control based on differing resource appropriations and stakeholder interests. We conducted an evaluation to determine if policy was associated with state-level 1) presence of wild pigs, 2) spatial extent of wild pig population occupancy, and 3) trajectory of wild pig population occupancy. Our results suggest the presence of wild pigs in various states was influenced by hunting preserves and the sale of hunting opportunities. In occupied states, the spatial extent of wild pigs was again associated with the sale of hunting opportunities and a wild pig hunting culture. Finally, the trajectory of state-level wild pig spatial occupancy was positively influenced by the sale of hunting opportunities, and negatively influenced by transportation policies. Based on these findings, we propose state governments standardize transportation policy and fenced hunting regulations across regions of the US in a more prohibitive fashion to diminish range expansion through illegal and negligent introductions via transportation, release, and escapes from game farms. Moreover, in states where wild pigs have yet to establish, we strongly recommend states proactively prohibit transportation through intra- and interstate movement.


Subject(s)
Animals, Wild , Introduced Species , Animals , United States , Swine , Agriculture , Farms , Sus scrofa
4.
Acta Biomater ; 121: 275-287, 2021 02.
Article in English | MEDLINE | ID: mdl-33129986

ABSTRACT

Traditional Anterior Cruciate Ligament (ACL) reconstruction is commonly performed using an allograft or autograft and possesses limitations such as donor site morbidity, decreased range of motion, and potential infection. However, a biodegradable synthetic graft could greatly assist in the prevention of such restrictions after ACL reconstruction. In this study, artificial grafts were generated using "wet" and "dry" electrospinning processes with a biodegradable elastomer, poly (ester urethane) urea (PEUU), and were evaluated in vitro and in vivo in a rat model. Four groups were established: (1) Wet PEUU artificial ligament, (2) Dry PEUU artificial ligament, (3) Dry polycaprolactone artificial ligament (PCL), and (4) autologous flexor digitorum longus tendon graft. Eight weeks after surgery, the in vivo tensile strength of wet PEUU ligaments had significantly increased compared to the other synthetic ligaments. These results aligned with increased infiltration of host cells and decreased inflammation within the wet PEUU grafts. In contrast, very little cellular infiltration was observed in PCL and dry PEUU grafts. Micro-computed tomography analysis performed at 4 and 8 weeks postoperatively revealed significantly smaller bone tunnels in the tendon autograft and wet PEUU groups. The Wet PEUU grafts served as an adequate functioning material and allowed for the creation of tissues that closely resembled the ACL.


Subject(s)
Anterior Cruciate Ligament Injuries , Anterior Cruciate Ligament Reconstruction , Animals , Anterior Cruciate Ligament/surgery , Anterior Cruciate Ligament Injuries/surgery , Rats , Tendons/surgery , Transplantation, Autologous , X-Ray Microtomography
5.
Mol Ther Methods Clin Dev ; 14: 64-76, 2019 Sep 13.
Article in English | MEDLINE | ID: mdl-31312666

ABSTRACT

Aging-related loss of adult stem cell function contributes to impaired tissue regeneration. Mice deficient in zinc metalloproteinase STE24 (Zmpste24 -/-) exhibit premature age-related musculoskeletal pathologies similar to those observed in children with Hutchinson-Gilford progeria syndrome (HGPS). We have reported that muscle-derived stem/progenitor cells (MDSPCs) isolated from Zmpste24 -/- mice are defective in their proliferation and differentiation capabilities in culture and during tissue regeneration. The mechanistic target of rapamycin complex 1 (mTORC1) regulates cell growth, and inhibition of the mTORC1 pathway extends the lifespan of several animal species. We therefore hypothesized that inhibition of mTORC1 signaling would rescue the differentiation defects observed in progeroid MDSPCs. MDSPCs were isolated from Zmpste24 -/- mice, and the effects of mTORC1 on MDSPC differentiation and function were examined. We found that mTORC1 signaling was increased in senescent Zmpste24 -/- MDSPCs, along with impaired chondrogenic, osteogenic, and myogenic differentiation capacity versus wild-type MDSPCs. Interestingly, we observed that mTORC1 inhibition with rapamycin improved myogenic and chondrogenic differentiation and reduced levels of apoptosis and senescence in Zmpste24 -/- MDSPCs. Our results demonstrate that age-related adult stem/progenitor cell dysfunction contributes to impaired regenerative capacities and that mTORC1 inhibition may represent a potential therapeutic strategy for improving differentiation capacities of senescent stem and muscle progenitor cells.

7.
J Orthop Res ; 35(7): 1375-1382, 2017 07.
Article in English | MEDLINE | ID: mdl-27572850

ABSTRACT

Mice expressing reduced levels of ERCC1-XPF (Ercc1-/Δ mice) demonstrate premature onset of age-related changes due to decreased repair of DNA damage. Muscle-derived stem/progenitor cells (MDSPCs) isolated from Ercc1-/Δ mice have an impaired capacity for cell differentiation. The mammalian target of rapamycin (mTOR) is a critical regulator of cell growth in response to nutrient, hormone, and oxygen levels. Inhibition of the mTOR pathway extends the lifespan of several species. Here, we examined the role of mTOR in regulating the MDSPC dysfunction that occurs with accelerated aging. We show that mTOR signaling pathways are activated in Ercc1-/Δ MDSPCs compared with wild-type (WT) MDSPCs. Additionally, inhibiting mTOR with rapamycin promoted autophagy and improved the myogenic differentiation capacity of the Ercc1-/Δ MDSPCs. The percent of apoptotic and senescent cells in Ercc1-/Δ MDSPC cultures was decreased upon mTOR inhibition. These results establish that mTOR signaling contributes to stem cell dysfunction and cell fate decisions in response to endogenous DNA damage. Therefore, mTOR represents a potential therapeutic target for improving defective, aged stem cells. © 2016 The Authors. Journal of Orthopaedic Research Published by Wiley Periodicals, Inc. on behalf of Orthopaedic Research Society. J Orthop Res 35:1375-1382, 2017.


Subject(s)
Aging/physiology , Multipotent Stem Cells/physiology , Progeria/physiopathology , TOR Serine-Threonine Kinases/metabolism , Animals , Apoptosis , Autophagy , Cell Differentiation , DNA-Binding Proteins/genetics , Disease Models, Animal , Endonucleases/genetics , Mice , Muscle Development , Progeria/genetics , Progeria/metabolism , Signal Transduction , Sirolimus , TOR Serine-Threonine Kinases/antagonists & inhibitors
8.
Am J Sports Med ; 45(3): 584-597, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27903590

ABSTRACT

BACKGROUND: Strong graft-bone integration is a prerequisite for successful graft remodeling after reconstruction of the anterior cruciate ligament (ACL) using soft tissue grafts. Novel strategies to accelerate soft tissue graft-bone integration are needed to reduce the need for bone-tendon-bone graft harvest, reduce patient convalescence, facilitate rehabilitation, and reduce total recovery time after ACL reconstruction. HYPOTHESIS: The application of ACL-derived stem cells with enhanced expression of bone morphogenetic protein 2 (BMP2) onto soft tissue grafts in the form of cell sheets will both accelerate and improve the quality of graft-bone integration after ACL reconstruction in a rat model. STUDY DESIGN: Controlled laboratory study. METHODS: ACL-derived CD34+ cells were isolated from remnant human ACL tissues, virally transduced to express BMP2, and embedded within cell sheets. In a rat model of ACL injury, bilateral single-bundle ACL reconstructions were performed, in which cell sheets were wrapped around tendon autografts before reconstruction. Four groups containing a total of 48 rats (96 knees) were established (n = 12 rats; 24 knees per group): CD34+BMP2 (100%), CD34+BMP2 (25%), CD34+ (untransduced), and a control group containing no cells. Six rats from each group were euthanized 2 and 4 weeks after surgery, and each graft was harvested for immunohistochemical and histological analyses. The remaining 6 rats in each group were euthanized at 4 and 8 weeks to evaluate in situ tensile load to failure in each femur-graft-tibia complex. RESULTS: In vitro, BMP2 transduction promoted the osteogenic differentiation of ACL-derived CD34+ cells while retaining their intrinsic multipotent capabilities. Osteoblast densities were greatest in the BMP2 (100%) and BMP2 (25%) groups. Bone tunnels in the CD34+BMP2 (100%) and CD34+BMP2 (25%) groups had the smallest cross-sectional areas according to micro-computed tomography analyses. Graft-bone integration occurred most rapidly in the CD34+BMP2 (25%) group. Tensile load to failure was significantly greater in the groups containing stem cells at 4 and 8 weeks after surgery. Tensile strength was greatest in the CD34+BMP2 (100%) group at 4 weeks, and in the CD34+BMP2 (25%) group at 8 weeks. CONCLUSION: ACL-derived CD34+ cells transduced with BMP2 accelerated graft-bone integration after ACL reconstruction using soft tissue autografts in a rat model, as evidenced by improved histological appearance and graft-bone interface biology along with tensile load to failure at each time point up to 8 weeks after surgery. CLINICAL RELEVANCE: A primary disadvantage of using soft tissue grafts for ACL reconstruction is the prolonged time required for bony ingrowth, which delays the initiation of midsubstance graft remodeling. The lack of consistent correlation between the appearance of a "healed" ACL on postoperative magnetic resonance imaging and readiness to return to sport results in athletes being released to sport before the graft is ready to handle high-intensity loading. Therefore, it is desirable to identify strategies that accelerate graft-bone integration, which would reduce the time to biologic fixation, improve the reliability of biologic fixation, allow for accelerated rehabilitation, and potentially reduce the incidence of early graft pullout and late midsubstance failure.


Subject(s)
Anterior Cruciate Ligament Reconstruction/methods , Anterior Cruciate Ligament/cytology , Bone Morphogenetic Protein 2/metabolism , Osteogenesis , Stem Cell Transplantation , Adolescent , Adult , Animals , Cell Differentiation , Female , Humans , Rats , Rats, Nude , Reproducibility of Results , Young Adult
9.
Hum Mol Genet ; 25(15): 3216-3231, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27354351

ABSTRACT

This study investigated the role of cyclooxygenase-2 (COX-2) expression by donor and host cells in muscle-derived stem cell (MDSC)-mediated bone regeneration utilizing a critical size calvarial defect model. We found that BMP4/green fluorescent protein (GFP)-transduced MDSCs formed significantly less bone in COX-2 knock-out (Cox-2KO) than in COX-2 wild-type (WT) mice. BMP4/GFP-transduced Cox-2KO MDSCs also formed significantly less bone than transduced WT MDSCs when transplanted into calvarial defects created in CD-1 nude mice. The impaired bone regeneration in the Cox-2KO MDSCBMP4/GFP group is associated with downregulation of BMP4-pSMAD1/5 signaling, decreased osteogenic differentiation and lowered proliferation capacity after transplantation, compared with WT MDSCBMP4/GFP cells. The Cox-2KO MDSCBMP4/GFP group demonstrated a reduction in cell survival and direct osteogenic differentiation in vitro These effects were mediated in part by the downregulation of Igf1 and Igf2. In addition, the Cox-2KO MDSCBMP4/GFP cells recruited fewer macrophages than the WT MDSC/BMP4/GFP cells in the early phase after injury. We concluded that the bone regeneration capacity of Cox-2KO MDSCs was impaired because of a reduction in cell proliferation and survival capacities, reduction in osteogenic differentiation and a decrease in the ability of the cells to recruit host cells to the injury site.


Subject(s)
Bone Regeneration , Cell Differentiation , Cell Proliferation , Cyclooxygenase 2/metabolism , Myoblasts , Skull/injuries , Stem Cell Transplantation , Animals , Cyclooxygenase 2/genetics , Mice , Mice, Knockout , Mice, Nude , Myoblasts/metabolism , Myoblasts/transplantation
10.
Biomaterials ; 60: 9-19, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25965282

ABSTRACT

Ruptured human anterior cruciate ligaments (ACL) contain vascular stem cells capable of enhancing the healing of tendon grafts. In the current study we explored the role that neo-angiogenesis plays in ACL healing. ACL-derived CD34+ cells were isolated via Fluorescence Activated Cell Sorting (FACS) from the rupture sites of human ACLs. The cells were then virally transduced to express either vascular endothelial growth factor (VEGF) or soluble FLT-1 (sFLT-1), which is an antagonist of VEGF. We established five groups: CD34+VEGF(100%), where 100% of the cells were transduced with VEGF, CD34+VEGF(25%), where only 25% of the cells were transduced with VEGF, CD34+, CD34+sFLT-1, and a No cells group. The CD34+sFLT1 group had a significant reduction in biomechanical strength compared to the CD34+ group at 4 and 8 weeks; whereas the biomechanical strength of the CD34+VEGF(25%) group was significantly greater than the CD34+ group at week 4; however, no difference was observed by week 8. Immunohistochemical staining demonstrated a significantly lower number of isolectin B4 and hCD31 positive cells, markers associated with angiogenesis, in the CD34+sFLT1 group, and a higher number of isolectin B4 and hCD31 positive cells in the CD34+VEGF(100%) and CD34+VEGF(25%) groups compared to the CD34+ group. Graft maturation was significantly delayed in the CD34+sFLT1 group and accelerated in the CD34+VEGF(25%) group compared to the CD34+ group. In conclusion, blocking VEGF reduced angiogenesis, graft maturation and biomechanical strength following ACL reconstruction. Native expression of VEGF by the CD34+ cells improved tendon graft maturation and biomechanical strength; however, over-expression of VEGF impeded improvements in biomechanical strength.


Subject(s)
Adult Stem Cells/transplantation , Anterior Cruciate Ligament Injuries , Anterior Cruciate Ligament Reconstruction/methods , Anterior Cruciate Ligament/blood supply , Neovascularization, Physiologic , Stem Cell Transplantation/methods , Adult , Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Animals , Anterior Cruciate Ligament/cytology , Anterior Cruciate Ligament/pathology , Antigens, CD34/analysis , Biomechanical Phenomena , Cells, Cultured , Gene Expression , Humans , Rats , Tissue Engineering , Transduction, Genetic , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism , Wound Healing
11.
Arthritis Res Ther ; 16(6): 482, 2014 Nov 17.
Article in English | MEDLINE | ID: mdl-25403236

ABSTRACT

INTRODUCTION: Recent studies have revealed that rapamycin activates autophagy in human chondrocytes preventing the development of osteoarthritis (OA) like changes in vitro, while the systemic injection of rapamycin reduces the severity of experimental osteoarthritis in a murine model of OA in vivo. Since the systemic use of rapamycin is associated with numerous side effects, the goal of the current study was to examine the beneficial effect of local intra-articular injection of rapamycin in a murine model of OA and to elucidate the mechanism of action of rapamycin on articular cartilage. METHODS: Destabilization of the medial meniscus (DMM) was performed on 10-week-old male mice to induce OA. Intra-articular injections of 10 µl of rapamycin (10 µM) were administered twice weekly for 8 weeks. Articular cartilage damage was analyzed by histology using a semi-quantitative scoring system at 8 and 12 weeks after surgery. Mammalian target of rapamycin (mTOR), light chain 3 (LC3), vascular endothelial growth factor (VEGF), collagen, type X alpha 1 (COL10A1), and matrix metallopeptidase 13 (MMP13) expressions were analyzed by immunohistochemistry. VEGF, COL10A1, and MMP13 expressions were further examined via quantitative RT-PCR (qPCR). RESULTS: Intra-articular injection of rapamycin significantly reduced the severity of articular cartilage degradation at 8 and 12 weeks after DMM surgery. A reduction in mTOR expression and the activation of LC3 (an autophagy marker) in the chondrocytes was observed in the rapamycin treated mice. Rapamycin treatment also reduced VEGF, COL10A1, and MMP13 expressions at 8 and 12 weeks after DMM surgery. CONCLUSION: These results demonstrate that the intra-articular injection of rapamycin could reduce mTOR expression, leading to a delay in articular cartilage degradation in our OA murine model. Our observations suggest that local intra-articular injection of rapamycin could represent a potential therapeutic approach to prevent OA.


Subject(s)
Cartilage, Articular/drug effects , Cartilage, Articular/pathology , Disease Models, Animal , Immunosuppressive Agents/administration & dosage , Osteoarthritis/drug therapy , Osteoarthritis/pathology , Sirolimus/administration & dosage , Animals , Cartilage, Articular/metabolism , Injections, Intra-Articular , Male , Mice , Osteoarthritis/metabolism
12.
J Orthop Res ; 32(10): 1326-32, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24964749

ABSTRACT

DNA damage is a cause of age related pathologies, including osteoarthritis (OA). Excision repair cross complementation group 1 (ERCC1) is an endonuclease required for DNA damage repair. In this study we investigated the function of ERCC1 in chondrocytes and its association with the pathophysiology of OA. ERCC1 expression in normal and osteoarthritic cartilage was assessed, as were changes in ERCC1 expression in chondrocytes under catabolic stress. Inhibiting ERCC1 in chondrocytes under interleukin-1ß stimulation using small interfering RNA (siRNA) was also evaluated. Finally, cellular senescence and apoptosis were examined in relation to ERCC1 function. ERCC1 expression was decreased in OA cartilage and increased within 4 h of exposure to interleukin (IL)-1ß, but decreased after 12 h. The inhibition of ERCC1 by siRNA increased the expression of matrix metallopeptidase 13 and decreased collagen type II. ERCC1 inhibition also increased the number of apoptotic and senescent cells. The inhibition of ERCC1 in chondrocytes increased their expression of OA related proteins, apoptosis, cellular senescence, and hypertrophic-like changes which suggest that ERCC1 is critical for protecting human chondrocytes (HCs) from catabolic stresses and provides insights into the pathophysiology of OA and a potential target for its treatment. (191)


Subject(s)
Apoptosis , Cellular Senescence , DNA-Binding Proteins/physiology , Endonucleases/physiology , Osteoarthritis/etiology , Osteoarthritis/physiopathology , Animals , Apoptosis/genetics , Cartilage, Articular/physiology , Cartilage, Articular/physiopathology , Cellular Senescence/genetics , Chondrocytes/pathology , Chondrocytes/physiology , DNA-Binding Proteins/genetics , Disease Models, Animal , Endonucleases/genetics , Humans , Mice , Mice, Inbred C57BL , Osteoarthritis/genetics
13.
Biomaterials ; 35(25): 6859-70, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24856105

ABSTRACT

Adult multipotent stem cells have been isolated from a variety of human tissues including human skeletal muscle, which represent an easily accessible source of stem cells. It has been shown that human skeletal muscle-derived stem cells (hMDSCs) are muscle-derived mesenchymal stem cells capable of multipotent differentiation. Although hMDSCs can undergo osteogenic differentiation and form bone when genetically modified to express BMP2; it is still unclear whether hMDSCs are as efficient as human bone marrow mesenchymal stem cells (hBMMSCs) for bone regeneration. The current study aimed to address this question by performing a parallel comparison between hMDSCs and hBMMSCs to evaluate their osteogenic and bone regeneration capacities. Our results demonstrated that hMDSCs and hBMMSCs had similar osteogenic-related gene expression profiles and had similar osteogenic differentiation capacities in vitro when transduced to express BMP2. Both the untransduced hMDSCs and hBMMSCs formed very negligible amounts of bone in the critical sized bone defect model when using a fibrin sealant scaffold; however, when genetically modified with lenti-BMP2, both populations successfully regenerated bone in the defect area. No significant differences were found in the newly formed bone volumes and bone defect coverage between the hMDSC and hBMMSC groups. Although both cell types formed mature bone tissue by 6 weeks post-implantation, the newly formed bone in the hMDSCs group underwent quicker remodelling than the hBMMSCs group. In conclusion, our results demonstrated that hMDSCs are as efficient as hBMMSCs in terms of their bone regeneration capacity; however, both cell types required genetic modification with BMP in order to regenerate bone in vivo.


Subject(s)
Bone Morphogenetic Protein 2/genetics , Bone Regeneration/physiology , Mesenchymal Stem Cells/metabolism , Muscle Cells/metabolism , Adult , Bone Morphogenetic Protein 2/metabolism , Bone and Bones/metabolism , Cell Differentiation/physiology , Cells, Cultured , Female , Genetic Vectors , Humans , Lentivirus/genetics , Male , Multipotent Stem Cells/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Osteogenesis/physiology , Tissue Engineering , Transcriptome , Transduction, Genetic , Young Adult
14.
FASEB J ; 28(8): 3792-809, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24843069

ABSTRACT

Murine muscle-derived stem cells (MDSCs) have been shown capable of regenerating bone in a critical size calvarial defect model when transduced with BMP 2 or 4; however, the contribution of the donor cells and their interactions with the host cells during the bone healing process have not been fully elucidated. To address this question, C57/BL/6J mice were divided into MDSC/BMP4/GFP, MDSC/GFP, and scaffold groups. After transplanting MDSCs into the critical-size calvarial defects created in normal mice, we found that mice transplanted with BMP4GFP-transduced MDSCs healed the bone defect in 4 wk, while the control groups (MDSC-GFP and scaffold) demonstrated no bone healing. The newly formed trabecular bone displayed similar biomechanical properties as the native bone, and the donor cells directly participated in endochondral bone formation via their differentiation into chondrocytes, osteoblasts, and osteocytes via the BMP4-pSMAD5 and COX-2-PGE2 signaling pathways. In contrast to the scaffold group, the MDSC groups attracted more inflammatory cells initially and incurred faster inflammation resolution, enhanced angiogenesis, and suppressed initial immune responses in the host mice. MDSCs were shown to attract macrophages via the secretion of monocyte chemotactic protein 1 and promote endothelial cell proliferation by secreting multiple growth factors. Our findings indicated that BMP4GFP-transduced MDSCs not only regenerated bone by direct differentiation, but also positively influenced the host cells to coordinate and promote bone tissue repair through paracrine effects.


Subject(s)
Bone Regeneration/physiology , Mesenchymal Stem Cell Transplantation , Animals , Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein 4/physiology , Cell Differentiation , Cell Movement , Chemokine CCL2/metabolism , Chondrocytes/cytology , Craniocerebral Trauma/surgery , Cyclooxygenase 2/physiology , Dinoprostone/physiology , Genes, Reporter , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/genetics , Inflammation , Intercellular Signaling Peptides and Proteins/metabolism , Macrophages , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/cytology , Neovascularization, Physiologic , Osteoblasts/cytology , Osteocytes/cytology , Paracrine Communication , Parietal Bone/injuries , Parietal Bone/physiology , Recombinant Fusion Proteins/metabolism , Signal Transduction , Smad5 Protein/physiology
16.
PLoS One ; 8(6): e64923, 2013.
Article in English | MEDLINE | ID: mdl-23762264

ABSTRACT

Human muscle-derived progenitor cells (hMDPCs) offer great promise for muscle cell-based regenerative medicine; however, prolonged ex-vivo expansion using animal sera is necessary to acquire sufficient cells for transplantation. Due to the risks associated with the use of animal sera, the development of a strategy for the ex vivo expansion of hMDPCs is required. The purpose of this study was to investigate the efficacy of using platelet-rich plasma (PRP) for the ex-vivo expansion of hMDPCs. Pre-plated MDPCs, myoendothelial cells, and pericytes are three populations of hMDPCs that we isolated by the modified pre-plate technique and Fluorescence Activated Cell Sorting (FACS), respectively. Pooled allogeneic human PRP was obtained from a local blood bank, and the effect that thrombin-activated PRP-releasate supplemented media had on the ex-vivo expansion of the hMDPCs was tested against FBS supplemented media, both in vitro and in vivo. PRP significantly enhanced short and long-term cell proliferation, with or without FBS supplementation. Antibody-neutralization of PDGF significantly blocked the mitogenic/proliferative effects that PRP had on the hMDPCs. A more stable and sustained expression of markers associated with stemness, and a decreased expression of lineage specific markers was observed in the PRP-expanded cells when compared with the FBS-expanded cells. The in vitro osteogenic, chondrogenic, and myogenic differentiation capacities of the hMDPCs were not altered when expanded in media supplemented with PRP. All populations of hMDPCs that were expanded in PRP supplemented media retained their ability to regenerate myofibers in vivo. Our data demonstrated that PRP promoted the proliferation and maintained the multi-differentiation capacities of the hMDPCs during ex-vivo expansion by maintaining the cells in an undifferentiated state. Moreover, PDGF appears to be a key contributing factor to the beneficial effect that PRP has on the proliferation of hMDPCs.


Subject(s)
Muscle Cells/cytology , Platelet-Rich Plasma/metabolism , Stem Cells/cytology , Aged , Animals , Biomarkers/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Flow Cytometry , Humans , Male , Mice , Mice, SCID , Middle Aged , Mitogens/pharmacology , Muscle Cells/drug effects , Muscle Cells/metabolism , Muscle Development/drug effects , Platelet-Derived Growth Factor/metabolism , Stem Cells/drug effects , Stem Cells/metabolism
17.
AIDS Res Hum Retroviruses ; 29(11): 1418-27, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23614610

ABSTRACT

Thirty years after its beginning, the HIV/AIDS epidemic is still raging around the world. According to UNAIDS, in 2011 alone 1.7M deaths were attributable to AIDS, and 2.5M people were newly infected by the virus. Despite the success in treating HIV-infected people with potent antiretroviral drugs, preventing HIV infection is the key to ending the epidemic. Recently, the efficacy of topical and systemic antiviral chemoprophylaxis (i.e., preexposure prophylaxis or "PrEP"), using the same drugs used for HIV treatment, has been demonstrated in a number of clinical trials. However, results from other trials have been inconsistent, especially those evaluating PrEP in women. These inconsistencies may result from our incomplete understanding of pharmacokinetics (PK)/pharmacodynamics (PD) at the mucosal sites of sexual transmission: the male and female gastrointestinal and reproductive tracts. The drug concentrations used in these trials were derived from those used for treatment; however, we still do not know the relationship between the therapeutic and the preventive dose. This article presents the first comprehensive review of the available data in the HIV pharmacology field from animal models to human studies, and outlines gaps, challenges, and future directions. Addressing these pharmacological gaps and challenges will be critical in selecting and advancing future PrEP candidates and strategies with the greatest impact on the HIV epidemic.


Subject(s)
Anti-HIV Agents/pharmacology , Anti-HIV Agents/pharmacokinetics , Chemoprevention/methods , Disease Transmission, Infectious/prevention & control , HIV Infections/prevention & control , Animals , Gastrointestinal Tract/chemistry , Genitalia/chemistry , Humans , Mucous Membrane/chemistry
18.
J Appl Physiol (1985) ; 114(2): 262-73, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23154994

ABSTRACT

Losartan (Los) is a Food and Drug Administration-approved antihypertensive medication that has a well-tolerated side effect profile. We have demonstrated that treatment with Los immediately after injury was effective at promoting muscle healing and inducing an antifibrotic effect in a murine model of skeletal muscle injury. We initially investigated the minimum effective dose of Los administration immediately after injury and subsequently determined whether the timing of administering a clinically relevant dose of Los would influence its effectiveness at improving muscle healing after muscle injury. In the first part of this study, mice were administered 3, 10, 30, or 300 mg·kg(-1)·day(-1) of Los immediately after injury, and the healing process was evaluated histologically and physiologically 4 wk after injury. In the second study, the clinically relevant dose of 10 mg·kg(-1)·day(-1) was administered immediately or started at 3 or 7 days postinjury. The administration of 300 mg·kg(-1)·day(-1) immediately following injury led to a significant increase in muscle regeneration, a significant decrease in fibrosis, and an improvement in muscle function. Moreover, we observed a significant decrease in fibrosis and a significant increase in muscle regeneration at 4 wk postinjury, when the clinically relevant dose of 10 mg·kg(-1)·day(-1) was administered at 3 or 7 days postinjury. Functional evaluation also demonstrated a significant improvement compared with the injured untreated control when Los treatment was initiated 3 days after injury. Our study revealed accelerated muscle healing when the 300 mg·kg(-1)·day(-1) of Los was administered immediately after injury and a clinically relevant dose of 10 mg·kg(-1)·day(-1) of Los was administered at 3 or 7 days postinjury.


Subject(s)
Antihypertensive Agents/pharmacology , Contusions/physiopathology , Losartan/pharmacology , Muscle, Skeletal/injuries , Wound Healing/drug effects , Animals , Cell Line , Dose-Response Relationship, Drug , Follistatin/metabolism , In Vitro Techniques , Mice , Mice, Inbred C57BL , Models, Animal , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Myoblasts/cytology , Myoblasts/drug effects , Myoblasts/metabolism , Myostatin/metabolism , Regeneration/drug effects , Regeneration/physiology , Time Factors , Wound Healing/physiology
19.
Cell Transplant ; 22(12): 2393-408, 2013.
Article in English | MEDLINE | ID: mdl-23244588

ABSTRACT

Muscle-derived cells have been successfully isolated using a variety of different methods and have been shown to possess multilineage differentiation capacities, including an ability to differentiate into articular cartilage and bone in vivo; however, the characterization of human muscle-derived stem cells (hMDSCs) and their bone regenerative capacities have not been fully investigated. Genetic modification of these cells may enhance their osteogenic capacity, which could potentially be applied to bone regenerative therapies. We found that hMDSCs, isolated by the preplate technique, consistently expressed the myogenic marker CD56, the pericyte/endothelial cell marker CD146, and the mesenchymal stem cell markers CD73, CD90, CD105, and CD44 but did not express the hematopoietic stem cell marker CD45, and they could undergo osteogenic, chondrogenic, adipogenic, and myogenic differentiation in vitro. In order to investigate the osteoinductive potential of hMDSCs, we constructed a retroviral vector expressing BMP4 and GFP and a lentiviral vector expressing BMP2. The BMP4-expressing hMDSCs were able to undergo osteogenic differentiation in vitro and exhibited enhanced mineralization compared to nontransduced cells; however, when transplanted into a calvarial defect, they failed to regenerate bone. Local administration of BMP4 protein and cell pretreatment with N-acetylcysteine (NAC), which improves cell survival, did not enhance the osteogenic capacity of the retro-BMP4-transduced cells. In contrast, lenti-BMP2-transduced hMDSCs not only exhibited enhanced in vitro osteogenic differentiation but also induced robust bone formation and nearly completely healed a critical-sized calvarial defect in CD-1 nude mice 6 weeks following transplantation. Herovici's staining of the regenerated bone demonstrated that the bone matrix contained a large amount of type I collagen. Our findings indicated that the hMDSCs are likely mesenchymal stem cells of muscle origin and that BMP2 is more efficient than BMP4 in promoting the bone regenerative capacity of the hMDSCs in vivo.


Subject(s)
Bone Diseases/surgery , Bone Morphogenetic Protein 4/metabolism , Bone Regeneration , Muscle, Skeletal/cytology , Stem Cells/cytology , Acetylcysteine/pharmacology , Adult , Aged, 80 and over , Animals , Biomarkers/metabolism , Bone Diseases/metabolism , Bone Diseases/pathology , Cell Survival/drug effects , Cells, Cultured , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Nude , Mice, SCID , Muscle, Skeletal/metabolism , Osteogenesis , Stem Cell Transplantation , Stem Cells/drug effects , Stem Cells/metabolism , Transplantation, Heterologous , Young Adult
20.
Stem Cell Res Ther ; 2(4): 31, 2011 Jul 08.
Article in English | MEDLINE | ID: mdl-21745421

ABSTRACT

We have found that when muscle-derived stem cells (MDSCs) are implanted into a variety of tissues only a small fraction of the donor cells can be found within the regenerated tissues and the vast majority of cells are host derived. This observation has also been documented by other investigators using a variety of different stem cell types. It is speculated that the transplanted stem cells release factors that modulate repair indirectly by mobilizing the host's cells and attracting them to the injury site in a paracrine manner. This process is loosely called a 'paracrine mechanism', but its effects are not necessarily restricted to the injury site. In support of this speculation, it has been reported that increasing angiogenesis leads to an improvement of cardiac function, while inhibiting angiogenesis reduces the regeneration capacity of the stem cells in the injured vascularized tissues. This observation supports the finding that most of the cells that contribute to the repair process are indeed chemo-attracted to the injury site, potentially through host neo-angiogenesis. Since it has recently been observed that cells residing within the walls of blood vessels (endothelial cells and pericytes) appear to represent an origin for post-natal stem cells, it is tempting to hypothesize that the promotion of tissue repair, via neo-angiogenesis, involves these blood vessel-derived stem cells. For non-vascularized tissues, such as articular cartilage, the regenerative property of the injected stem cells still promotes a paracrine, or bystander, effect, which involves the resident cells found within the injured microenvironment, albeit not through the promotion of angiogenesis. In this paper, we review the current knowledge of post-natal stem cell therapy and demonstrate the influence that implanted stem cells have on the tissue regeneration and repair process. We argue that the terminal differentiation capacity of implanted stem cells is not the major determinant of the cells regenerative potential and that the paracrine effect imparted by the transplanted cells plays a greater role in the regeneration process.


Subject(s)
Cell Differentiation , Stem Cell Transplantation , Animals , Cartilage Diseases/pathology , Cartilage Diseases/therapy , Cell- and Tissue-Based Therapy , Cellular Microenvironment , Heart Diseases/pathology , Heart Diseases/therapy , Humans , Mice , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/physiology , Neovascularization, Pathologic , Paracrine Communication , Regeneration , Stem Cells/cytology , Stem Cells/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...