Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Cell Mol Med ; 22(12): 6015-6025, 2018 12.
Article in English | MEDLINE | ID: mdl-30216653

ABSTRACT

Mesenchymal stromal cell (MSC) therapies combined with renal pulsed focused ultrasound (pFUS) pretreatment increase MSC homing and improve cisplatin-induced acute kidney injury (AKI) better than MSC alone. However, mechanisms underlying improved outcomes remain unknown. We hypothesize pFUS up-regulates renal interferon-γ (IFNγ) and stimulates MSC to produce interleukin-10 (IL-10) after migrating to kidneys. To demonstrate initially, MSC cultured with IFNγ up-regulated IL-10. More MSC-derived IL-10 was detected in kidneys when IFNγ-stimulated MSC were infused and they improved AKI better than unstimulated MSC. Next, IFNγ-knockout mice with AKI received pFUS+MSC, but MSC-derived IL-10 expression and AKI were similar to using MSC alone. AKI in wild-type mice receiving pFUS and IL-10-deficient MSC was also unimproved compared to administering IL-10-deficient MSC alone. Indoleamine 2,3-dioxygenase (IDO), an anti-inflammatory enzyme up-regulated in MSC by IFNγ, was up-regulated during AKI, but was not further elevated in MSC from pFUS-treated kidneys, suggesting that IDO is not involved in improved AKI healing by pFUS+MSC. These data suggest IFNγ is up-regulated by pFUS and after i.v.-infused MSC home to pFUS-treated kidneys, IFNγ stimulates additional IL-10 production by MSC to improve AKI. Analogous mechanisms of ultrasound-treated tissue microenvironments stimulating therapeutic MSC may exist in other pathologies where adjuvant ultrasound techniques are successful.


Subject(s)
Acute Kidney Injury/therapy , Interferon-gamma/genetics , Interleukin-10/genetics , Mesenchymal Stem Cell Transplantation , Acute Kidney Injury/chemically induced , Acute Kidney Injury/genetics , Animals , Cisplatin/adverse effects , Disease Models, Animal , Gene Expression Regulation, Developmental/radiation effects , Humans , Kidney/injuries , Kidney/metabolism , Kidney/pathology , Mesenchymal Stem Cells , Mice , Mice, Knockout , Ultrasonic Waves
2.
Sci Rep ; 7(1): 8607, 2017 08 17.
Article in English | MEDLINE | ID: mdl-28819122

ABSTRACT

Mesenchymal stromal cells (MSC) are potential renal therapeutics. Clinically, results are mixed partly because MSC tropism to kidneys is minimal following infusion. Ultrasound augmentation of the renal microenvironment is becoming increasingly-important in renal MSC therapies. We demonstrated pulsed-focused-ultrasound (pFUS) increases enhanced homing permeability and retention of MSC in mouse kidneys. Here, we characterized the temporal proteomic response to pFUS in mouse kidneys and its association with MSC tropism. pFUS induced molecular cascades of initial increases in tumor necrosis factor-α (TNFα) and interleukin (IL)-1α, that activated nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB) and cyclooxygenase-2 (COX2) pathways without cell death. This was followed by a 24-48 hour-long response of increased cell adhesion molecules (CAM), trophic and anti-inflammatory factors. Pretreating animals with anti-inflammatory drugs etanercept (TNFα inhibitor), anakinra (IL-1 receptor antagonist), prednisone (NFκB translocation inhibitor), or ibuprofen (COX inhibitor) suppressed molecular changes and inhibited renal MSC tropism. We further examined the role of COX2 using a COX2-knock-out mouse where pFUS was unable to increase MSC tropism. These results demonstrate that renal micro-environmental changes induce MSC tropism and could influence the therapeutic efficacy of MSC. Optimizing the microenvironment and understanding drug effects will enable improvements in MSC therapies for renal disease.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Kidney/cytology , Kidney/diagnostic imaging , Mesenchymal Stem Cells/metabolism , Tropism , Ultrasonic Waves , Animals , Cyclooxygenase 2/metabolism , Female , Humans , Kidney/drug effects , Macrophages/metabolism , Mice, Knockout , Proteomics
3.
Sci Rep ; 7: 41550, 2017 02 07.
Article in English | MEDLINE | ID: mdl-28169278

ABSTRACT

Mesenchymal stem cells (MSC) are promising therapeutics for critical limb ischemia (CLI). Mechanotransduction from pulsed focused ultrasound (pFUS) upregulates local chemoattractants to enhance homing of intravenously (IV)-infused MSC and improve outcomes. This study investigated whether pFUS exposures to skeletal muscle would improve local homing of iv-infused MSCs and their therapeutic efficacy compared to iv-infused MSCs alone. CLI was induced by external iliac arterial cauterization in 10-12-month-old mice. pFUS/MSC treatments were delayed 14 days, when surgical inflammation subsided. Mice were treated with iv-saline, pFUS alone, IV-MSC, or pFUS and IV-MSC. Proteomic analyses revealed pFUS upregulated local chemoattractants and increased MSC tropism to CLI muscle. By 7 weeks post-treatment, pFUS + MSC significantly increased perfusion and CD31 expression, while reducing fibrosis compared to saline. pFUS or MSC alone reduced fibrosis, but did not increase perfusion or CD31. Furthermore, MSCs homing to pFUS-treated CLI muscle expressed more vascular endothelial growth factor (VEGF) and interleukin-10 (IL-10) than MSCs homing to non-pFUS-treated muscle. pFUS + MSC improved perfusion and vascular density in this clinically-relevant CLI model. The molecular effects of pFUS increased both MSC homing and MSC production of VEGF and IL-10, suggesting microenvironmental changes from pFUS also increased potency of MSCs in situ to further enhance their efficacy.


Subject(s)
Extremities/blood supply , Ischemia/pathology , Ischemia/physiopathology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Reperfusion , Ultrasonic Waves , Animals , Biomarkers , Disease Models, Animal , Extremities/radiation effects , Female , Fibrosis , Interleukin-10/genetics , Interleukin-10/metabolism , Ischemia/diagnostic imaging , Ischemia/therapy , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Muscle, Skeletal/blood supply , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Proteome , Proteomics/methods , Reperfusion/methods , Ultrasonography, Doppler, Color , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
4.
Nanomedicine ; 13(2): 503-513, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27520728

ABSTRACT

Stem cell-based therapies have become a major focus in regenerative medicine and to treat diseases. A straightforward approach combining three drugs, heparin (H), protamine (P) with ferumoxytol (F) in the form of nanocomplexes (NCs) effectively labeled stem cells for cellular MRI. We report on the physicochemical characteristics for optimizing the H, P, and F components in different ratios, and mixing sequences, producing NCs that varied in hydrodynamic size. NC size depended on the order in which drugs were mixed in media. Electron microscopy of HPF or FHP showed that F was located on the surface of spheroidal shaped HP complexes. Human stem cells incubated with FHP NCs resulted in a significantly greater iron concentration per cell compared to that found in HPF NCs with the same concentration of F. These results indicate that FHP could be useful for labeling stem cells in translational studies in the clinic.


Subject(s)
Ferrosoferric Oxide , Heparin , Protamines , Stem Cells , Cell Tracking , Humans , Magnetic Resonance Imaging , Magnetics , Nanoparticles , Stem Cell Transplantation
5.
J Neuroimmunol ; 292: 116-25, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26943969

ABSTRACT

The transgenic HIV-1 rat (Tg) is a commonly used neuroHIV model with documented neurologic/behavioral deficits. Using immunofluorescent staining of the Tg brain, we found astrocytic dysfunction/damage, as well as dopaminergic neuronal loss/dysfunction, both of which worsening significantly in the striatum with age. We saw mild microglial activation in young Tg brains, but this decreased with age. There were no differences in neurogenesis potential suggesting a neurodegenerative rather than a neurodevelopmental process. Gp120 CSF levels exceeded serum gp120 levels in some animals, suggesting local viral protein production in the brain. Further probing of the pathophysiology underlying astrocytic injury in this model is warranted.


Subject(s)
AIDS-Associated Nephropathy/pathology , AIDS-Associated Nephropathy/virology , Aging/pathology , Brain/pathology , Gene Expression Regulation, Viral/physiology , AIDS-Associated Nephropathy/metabolism , Animals , Brain/virology , CD11b Antigen/genetics , CD11b Antigen/metabolism , Calcium-Binding Proteins/metabolism , Case-Control Studies , Enzyme-Linked Immunosorbent Assay , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , HIV Envelope Protein gp120/cerebrospinal fluid , HIV Envelope Protein gp120/genetics , HIV-1/genetics , HIV-1/pathogenicity , Humans , Male , Microfilament Proteins/metabolism , Rats , Rats, Transgenic , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism
6.
Contrast Media Mol Imaging ; 11(1): 55-64, 2016.
Article in English | MEDLINE | ID: mdl-26234504

ABSTRACT

To develop effective stem cell therapies, it is important to track therapeutic cells non-invasively and monitor homing to areas of pathology. The purpose of this study was to design and evaluate the labeling efficiency of commercially available dextran-coated superparamagnetic iron oxide nanoparticles, FeraTrack Direct (FTD), in various stem and immune cells; assess the cytotoxicity and tolerability of the FTD in stem cells; and monitor stem cell homing using FTD-labeled bone-marrow-derived mesenchymal stromal cells (BMSCs) and neural stem cells (NSCs) in a tumor model by in vivo MRI. BMSCs, NSCs, hematopoietic stem cells (HSCs), T-lymphocytes, and monocytes were labeled effectively with FTD without the need for transfection agents, and Prussian blue (PB) staining and transmission electron microscopy (TEM) confirmed intracellular uptake of the agent. The viability, proliferation, and functionality of the labeled cells were minimally or not affected after labeling. When 10(6) FTD-labeled BMSCs or NSCs were injected into C6 glioma bearing nude mice, the cells homing to the tumors were detected as hypointense regions within the tumor using 3 T clinical MRI up to 10 days post injection. Histological analysis confirmed the homing of injected cells to the tumor by the presence of PB positive cells that are not macrophages. Labeling of stem cells or immune cells with FTD was non-toxic, and should facilitate the translation of this agent to clinical trials for evaluation of trafficking of cells by MRI.


Subject(s)
Cell Proliferation , Cell Tracking , Ferric Compounds/chemistry , Staining and Labeling , Animals , Hematopoietic Stem Cells/chemistry , Hematopoietic Stem Cells/ultrastructure , Magnetic Resonance Imaging , Magnetite Nanoparticles/chemistry , Mesenchymal Stem Cells/chemistry , Mesenchymal Stem Cells/ultrastructure , Mice , Microscopy, Electron, Transmission
7.
Stem Cells ; 33(4): 1241-53, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25640064

ABSTRACT

Animal studies have shown that mesenchymal stromal cell (MSC) infusions improve acute kidney injury (AKI) outcomes when administered early after ischemic/reperfusion injury or within 24 hours after cisplatin administration. These findings have spurred several human clinical trials to prevent AKI. However, no specific therapy effectively treats clinically obvious AKI or rescues renal function once advanced injury is established. We investigated if noninvasive image-guided pulsed focused ultrasound (pFUS) could alter the kidney microenvironment to enhance homing of subsequently infused MSC. To examine the efficacy of pFUS-enhanced cell homing in disease, we targeted pFUS to kidneys to enhance MSC homing after cisplatin-induced AKI. We found that pFUS enhanced MSC homing at 1 day post-cisplatin, prior to renal functional deficits, and that enhanced homing improved outcomes of renal function, tubular cell death, and regeneration at 5 days post-cisplatin compared to MSC alone. We then investigated whether pFUS+MSC therapy could rescue established AKI. MSC alone at 3 days post-cisplatin, after renal functional deficits were obvious, significantly improved 7-day survival of animals. Survival was further improved by pFUS and MSC. pFUS prior to MSC injections increased IL-10 production by MSC that homed to kidneys and generated an anti-inflammatory immune cell profile in treated kidneys. This study shows pFUS is a neoadjuvant approach to improve MSC homing to diseased organs. pFUS with MSC better prevents AKI than MSC alone and allows rescue therapy in established AKI, which currently has no meaningful therapeutic options.


Subject(s)
Acute Kidney Injury/therapy , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/physiology , Ultrasonic Waves , Acute Kidney Injury/pathology , Animals , Female , Humans , Mesenchymal Stem Cells/radiation effects , Mice , Mice, Inbred C3H , Treatment Outcome
8.
Stem Cells ; 33(4): 1173-86, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25534849

ABSTRACT

Maximal homing of infused stem cells to diseased tissue is critical for regenerative medicine. Pulsed focused ultrasound (pFUS) is a clinically relevant platform to direct stem cell migration. Through mechanotransduction, pFUS establishes local gradients of cytokines, chemokines, trophic factors (CCTF) and cell adhesion molecules (CAM) in treated skeletal muscle that subsequently infused mesenchymal stromal cells (MSC) can capitalize to migrate into the parenchyma. Characterizing molecular responses to mechanical pFUS effects revealed tumor necrosis factor-alpha (TNFα) drives cyclooxygenase-2 (COX2) signaling to locally increase CCTF/CAM that are necessary for MSC homing. pFUS failed to increase chemoattractants and induce MSC homing to treated muscle in mice pretreated with ibuprofen (nonspecific COX inhibitor) or etanercept (TNFα inhibitor). pFUS-induced MSC homing was also suppressed in COX2-knockout mice, demonstrating ibuprofen blocked the mechanically induced CCTF/CAM by acting on COX2. Anti-inflammatory drugs, including ibuprofen, are administered to muscular dystrophy (MD) patients, and ibuprofen also suppressed pFUS-induced homing to muscle in a mouse model of MD. Drug interactions with cell therapies remain unexplored and are not controlled for during clinical cell therapy trials. This study highlights potentially negative drug-host interactions that suppress stem cell homing and could undermine cell-based approaches for regenerative medicine.


Subject(s)
Cyclooxygenase 2 Inhibitors/pharmacology , Mechanotransduction, Cellular/physiology , Mesenchymal Stem Cells/physiology , Muscular Dystrophies , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Ultrasonic Waves , Animals , Cells, Cultured , Female , Humans , Mechanotransduction, Cellular/drug effects , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/radiation effects , Mice , Mice, 129 Strain , Mice, Inbred C3H , Mice, Knockout , Muscular Dystrophies/pathology , Tumor Necrosis Factor-alpha/biosynthesis , Young Adult
9.
Stem Cells ; 31(11): 2551-60, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23922277

ABSTRACT

Stem cells are promising therapeutics for cardiovascular diseases, and i.v. injection is the most desirable route of administration clinically. Subsequent homing of exogenous stem cells to pathological loci is frequently required for therapeutic efficacy and is mediated by chemoattractants (cell adhesion molecules, cytokines, and growth factors). Homing processes are inefficient and depend on short-lived pathological inflammation that limits the window of opportunity for cell injections. Noninvasive pulsed focused ultrasound (pFUS), which emphasizes mechanical ultrasound-tissue interactions, can be precisely targeted in the body and is a promising approach to target and maximize stem cell delivery by stimulating chemoattractant expression in pFUS-treated tissue prior to cell infusions. We demonstrate that pFUS is nondestructive to murine skeletal muscle tissue (no necrosis, hemorrhage, or muscle stem cell activation) and initiates a largely M2-type macrophage response. We also demonstrate that local upregulation of chemoattractants in pFUS-treated skeletal muscle leads to enhance homing, permeability, and retention of human mesenchymal stem cells (MSC) and human endothelial precursor cells (EPC). Furthermore, the magnitude of MSC or EPC homing was increased when pFUS treatments and cell infusions were repeated daily. This study demonstrates that pFUS defines transient "molecular zip codes" of elevated chemoattractants in targeted muscle tissue, which effectively provides spatiotemporal control and tunability of the homing process for multiple stem cell types. pFUS is a clinically translatable modality that may ultimately improve homing efficiency and flexibility of cell therapies for cardiovascular diseases.


Subject(s)
Cytokines/metabolism , Mesenchymal Stem Cells/metabolism , Muscle, Skeletal/metabolism , Stem Cells/metabolism , Ultrasonics/methods , Animals , Cell Culture Techniques , Female , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/diagnostic imaging , Mice , Mice, Inbred C3H , Models, Animal , Muscle, Skeletal/cytology , Muscle, Skeletal/diagnostic imaging , Spatio-Temporal Analysis , Stem Cells/cytology , Stem Cells/diagnostic imaging , Ultrasonography
SELECTION OF CITATIONS
SEARCH DETAIL
...