Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
2.
Transl Vis Sci Technol ; 10(10): 3, 2021 08 12.
Article in English | MEDLINE | ID: mdl-34383879

ABSTRACT

Purpose: Mesenchymal stromal cells (MSCs) have been shown to enhance tissue repair as a cell-based therapy. In preparation for a phase I clinical study, we evaluated the safety, dosing, and efficacy of bone marrow-derived MSCs after subconjunctival injection in preclinical animal models of mice, rats, and rabbits. Methods: Human bone marrow-derived MSCs were expanded to passage 4 and cryopreserved. Viability of MSCs after thawing and injection through small-gauge needles was evaluated by vital dye staining. The in vivo safety of human and rabbit MSCs was studied by subconjunctivally injecting MSCs in rabbits with follow-up to 90 days. The potency of MSCs on accelerating wound healing was evaluated in vitro using a scratch assay and in vivo using 2-mm corneal epithelial debridement wounds in mice. Human MSCs were tracked after subconjunctival injection in rat and rabbit eyes. Results: The viability of MSCs after thawing and immediate injection through 27- and 30-gauge needles was 93.1% ± 2.1% and 94.9% ± 1.3%, respectively. Rabbit eyes demonstrated mild self-limiting conjunctival inflammation at the site of injection with human but not rabbit MSCs. In scratch assay, the mean wound healing area was 93.5% ± 12.1% in epithelial cells co-cultured with MSCs compared with 40.8% ± 23.1% in controls. At 24 hours after wounding, all MSC-injected murine eyes had 100% corneal wound closure compared with 79.9% ± 5.5% in controls. Human MSCs were detectable in the subconjunctival area and peripheral cornea at 14 days after injection. Conclusions: Subconjunctival administration of MSCs is safe and effective in promoting corneal epithelial wound healing in animal models. Translational Relevance: These results provide preclinical data to support a phase I clinical study.


Subject(s)
Corneal Injuries , Mesenchymal Stem Cells , Animals , Bone Marrow , Clinical Trials, Phase I as Topic , Cornea , Corneal Injuries/therapy , Mice , Rabbits , Rats , Wound Healing
3.
Sci Transl Med ; 12(573)2020 12 09.
Article in English | MEDLINE | ID: mdl-33298563

ABSTRACT

In autosomal dominant conditions with haploinsufficiency, a single functional allele cannot maintain sufficient dosage for normal function. We hypothesized that pharmacologic induction of the wild-type allele could lead to gene dosage compensation and mitigation of the disease manifestations. The paired box 6 (PAX6) gene is crucial in tissue development and maintenance particularly in eye, brain, and pancreas. Aniridia is a panocular condition with impaired eye development and limited vision due to PAX6 haploinsufficiency. To test our hypothesis, we performed a chemical screen and found mitogen-activated protein kinase kinase (MEK) inhibitors to induce PAX6 expression in normal and mutant corneal cells. Treatment of newborn Pax6-deficient mice (Pax6Sey-Neu/+ ) with topical or systemic MEK inhibitor PD0325901 led to increased corneal PAX6 expression, improved corneal morphology, reduced corneal opacity, and enhanced ocular function. These results suggest that induction of the wild-type allele by drug repurposing is a potential therapeutic strategy for haploinsufficiencies, which is not limited to specific mutations.


Subject(s)
Haploinsufficiency , Paired Box Transcription Factors , Animals , Eye Proteins/genetics , Gene Dosage , Homeodomain Proteins/genetics , Mice , PAX6 Transcription Factor/genetics , Paired Box Transcription Factors/genetics , Repressor Proteins/genetics
4.
Heliyon ; 6(10): e04984, 2020 Oct.
Article in English | MEDLINE | ID: mdl-33083585

ABSTRACT

Terpineol, a promising valorisation product of pine industry, is widely used as an active ingredient for disinfectant soap, cleansers, perfumes, and pharmaceutical purposes. Synthesis of terpineol is generally carried out by separation of α-pinene compounds from crude turpentine through fractionation and then hydrated (addition of water) with the help of acid catalysts. However, direct turpentine hydration without pre-fractionation process can be more beneficial from economic and process point of views. This study aims to investigate the effect of both single and mixed/combined catalysts towards terpineol yield. Combined strong and weak acid catalysts were required to obtain high feed conversion and terpineol yield. The selectivity of terpineol is then correlated to the solubility of a weak/organic acid. In this study, the highest yield of terpineol was 54.0 ± 8.2%-w/w using combination of formic acid and sulphuric acid.

5.
Transl Vis Sci Technol ; 9(3): 26, 2020 02 21.
Article in English | MEDLINE | ID: mdl-32742756

ABSTRACT

Purpose: A reproducible protocol for the production of corneal mesenchymal stem/stromal cells (cMSCs) is necessary for potential clinical applications. We aimed to describe successful generation and expansion of cMSCs using an explant method. Methods: Corneoscleral rims of human cadaveric eyes were divided into four pieces and used as explants to allow outgrowth of cMSCs (passage 0, or P0). The cells were subcultured at a 1:10 ratio until passage 5 (P5). The characteristics as well as therapeutic effects of expanded cMSCs were evaluated both in vitro, using a scratch assay, and in vivo using epithelial debridement and chemical injury mouse models. Results: All explants demonstrated outgrowth of cells by 7 days. Although the initial outgrowth included mixed mesenchymal and epithelial cells, by P1 only cMSCs remained. By subculturing each flask at a ratio of 1:10, the potential yield from each cornea was approximately 12 to 16 × 1010 P5 cells. P5 cMSCs demonstrated the cell surface markers of MSCs. The secretome of P5 cMSCs induced faster closure of wounds in an in vitro scratch assay. Subconjunctival injection of P5 cMSCs in mouse models of mechanical corneal epithelial debridement or ethanol injury led to significantly faster wound healing and decreased inflammation, relative to control. Conclusions: cMSCs can be reproducibly derived from human cadaveric corneas using an explant method and expanded with preservation of characteristics and corneal wound healing effects. Translational Relevance: The results of our study showed that cMSCs produced using this scheme can be potentially used for clinical applications.


Subject(s)
Burns, Chemical , Corneal Injuries , Mesenchymal Stem Cells , Animals , Cornea , Corneal Injuries/therapy , Wound Healing
6.
Exp Eye Res ; 181: 263-270, 2019 04.
Article in English | MEDLINE | ID: mdl-30822400

ABSTRACT

Colonization by Staphylococcus aureus (S. aureus) has been implicated in many infectious and wound healing disorders. This study was performed to characterize the pathogenic role of S. aureus alpha-hemolysin (alpha-toxin) in corneal epithelial wound healing and infectious keratitis in the setting of a corneal wound. The effect of wild-type and isogenic Hla mutant (α-hemolysin gene deleted) S. aureus bacteria and conditioned media on corneal epithelial wound healing was tested in vitro using a scratch assay and in vivo using a murine epithelial debridement model. The invasiveness of wild-type and Hla mutant S. aureus was evaluated in vitro in human corneal epithelial cells and in vivo in a murine model of infectious keratitis following total epithelial debridement. S. aureus and its conditioned media significantly delayed epithelial wound closure both in vitro (P < 0.05) and in vivo (P < 0.05). The effect of S. aureus on wound healing was significantly diminished with the Hla mutant strain (P < 0.05). Likewise, compared to the wild-type strain, the Hla mutant strain demonstrated significantly reduced ability to invade corneal epithelial cells in vitro (P < 0.05) and infect murine corneas following total epithelial debridement in vivo (P < 0.05). In conclusion, S. aureus alpha-hemolysin plays a major role in the pathologic modulation of corneal epithelial wound healing and the intracellular invasion of the bacteria. Limiting colonization by S. aureus and/or blocking alpha-hemolysin may provide a therapeutic approach for corneal wound healing and infectious disorders.


Subject(s)
Corneal Diseases/microbiology , Epithelium, Corneal/injuries , Hemolysin Proteins/physiology , Staphylococcal Infections/microbiology , Staphylococcus aureus/pathogenicity , Wound Healing/physiology , Animals , Corneal Diseases/pathology , Disease Models, Animal , Epithelial Cells/microbiology , Epithelium, Corneal/microbiology , Humans , Keratitis/microbiology , Mice , Mice, Inbred C57BL , Staphylococcal Infections/pathology
7.
Stem Cells Transl Med ; 8(5): 478-489, 2019 05.
Article in English | MEDLINE | ID: mdl-30644653

ABSTRACT

Severe corneal injuries often result in permanent vision loss and remain a clinical challenge. Human bone marrow-derived mesenchymal stem cells (MSCs) and their secreted factors (secretome) have been studied for their antiscarring, anti-inflammatory, and antiangiogeneic properties. We aimed to deliver lyophilized MSC secretome (MSC-S) within a viscoelastic gel composed of hyaluronic acid (HA) and chondroitin sulfate (CS) as a way to enhance corneal re-epithelialization and reduce complications after mechanical and chemical injuries of the cornea. We hypothesized that delivering MSC-S within HA/CS would have improved wound healing effects compared the with either MSC-S or HA/CS alone. The results showed that a once-daily application of MSC-S in HA/CS enhances epithelial cell proliferation and wound healing after injury to the cornea. It also reduced scar formation, neovascularization, and hemorrhage after alkaline corneal burns. We found that combining MSC-S and HA/CS increased the expression of CD44 receptors colocalized with HA, suggesting that the observed therapeutic effects between the MSC-S and HA/CS are in part mediated by CD44 receptor upregulation and activation by HA. The results from this study demonstrate a reproducible and efficient approach for delivering the MSC-S to the ocular surface for treatment of severe corneal injuries. Stem Cells Translational Medicine 2019;8:478-489.


Subject(s)
Cornea/pathology , Corneal Injuries/therapy , Mesenchymal Stem Cells/metabolism , Proteome/metabolism , Viscoelastic Substances/therapeutic use , Wound Healing/physiology , Animals , Female , Humans , Rats , Rats, Sprague-Dawley , Viscoelastic Substances/pharmacology
8.
J Cataract Refract Surg ; 45(3): 361-366, 2019 03.
Article in English | MEDLINE | ID: mdl-30527441

ABSTRACT

PURPOSE: To develop a reproducible ex vivo model of corneal endothelial cell injury using phacoemulsification in porcine eyes and to evaluate the effects of mesenchymal stromal cell secretome in this injury model. SETTING: Department of Ophthalmology, University of Illinois at Chicago, Illinois, USA. DESIGN: Experimental study. METHODS: A corneal endothelial injury model was optimized using different powers and durations of ultrasound energy inside ex vivo porcine eyes. Conditioned media from corneal mesenchymal stem cells was collected under serum-free conditions from passages 4 to 6. Immediately after the phacoemulsification injury, the anterior chamber fluid was replaced with unconditioned media or conditioned media and incubated at 37°C for 4 hours. At the end, endothelial cell viability was evaluated using trypan blue staining and analyzed with ImageJ software. RESULTS: Using specific parameters (50% power for 30 seconds), phacoemulsification inside fresh porcine eyes led to a consistent level of endothelial cell injury. Incubation with corneal mesenchymal stromal cell-conditioned media after the injury significantly reduced endothelial cells loss compared with unconditioned media (mean 1.29% ± 0.91% [SD] and 5.33% ± 3.24%, respectively, P < .05). CONCLUSIONS: Phacoemulsification inside fresh porcine eyes provided a reproducible model to study endothelial cell injury. Treatment with corneal mesenchymal stromal cell secretome after injury appeared to significantly enhance the survival of corneal endothelial cells. This might provide a new strategy for preventing corneal endothelial cell loss after phacoemulsification or other endothelial injuries. Further in vivo studies are necessary to determine the therapeutic potential.


Subject(s)
Corneal Endothelial Cell Loss/prevention & control , Endothelium, Corneal , Mesenchymal Stem Cells/metabolism , Metabolome/physiology , Phacoemulsification/adverse effects , Animals , Culture Media , Disease Models, Animal , Endothelium, Corneal/drug effects , Endothelium, Corneal/injuries , Swine
9.
Invest Ophthalmol Vis Sci ; 59(12): 5194-5200, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30372747

ABSTRACT

Purpose: Mesenchymal stromal cells (MSCs) have been used therapeutically to modulate inflammation and promote repair. Extracellular vesicles, including exosomes, have been identified as one of the important mediators. This study investigated the effect of human corneal MSC-derived exosomes on corneal epithelial wound healing. Methods: Corneal MSCs (cMSCs) were isolated from human cadaver corneas. The secretome was collected after 72 hours and exosomes were isolated using differential ultracentrifugation. Morphology and size of exosomes were examined by electron microscopy and dynamic light scattering. Expression of CD9, CD63, and CD81 by cMSC exosomes was evaluated by western blotting. Cellular uptake of exosomes was studied using calcein-stained exosomes. The effect of exosome on wound healing was measured in vitro using a scratch assay and in vivo after 2-mm epithelial debridement wounds in mice. Results: cMSC exosomes were morphologically round and main population ranged between 40 and 100 nm in diameter. They expressed CD9, CD63, and CD81, and did not express GM130, Calnexin, and Cytochrome-C. Stained cMSC exosomes were successfully taken up by human cMSCs, human corneal epithelial cells (HCECs), and human macrophages in vitro and by corneal epithelium in vivo. In scratch assay, after 16 hours, cMSC exosome treated HCECs had 30.1% ± 14% remaining wound area compared to 72.9% ± 8% in control (P < 0.005). In vivo, after 72 hours, cMSC exosome-treated corneas had 77.5% ± 3% corneal wound healing compared to 41.6% ± 7% in the control group (P < 0.05). Conclusions: Human cMSC exosomes can accelerate corneal epithelial wound healing, and thus, may provide a therapeutic approach for ocular surface injuries.


Subject(s)
Cornea/cytology , Corneal Injuries/physiopathology , Epithelium, Corneal/injuries , Exosomes/physiology , Mesenchymal Stem Cells/physiology , Wound Healing/physiology , Animals , Antigens, CD/metabolism , Blotting, Western , Cell Proliferation , Corneal Injuries/metabolism , Humans , Light , Mesenchymal Stem Cells/cytology , Mice , Mice, Inbred C57BL , Microscopy, Electron , Scattering, Radiation
10.
Stem Cells ; 36(5): 775-784, 2018 05.
Article in English | MEDLINE | ID: mdl-29341332

ABSTRACT

Macrophages are crucial drivers of inflammatory corneal neovascularization and thus are potential targets for immunomodulatory therapies. We hypothesized that therapeutic use of cornea-derived mesenchymal stromal cells (cMSCs) may alter the function of macrophages. We found that cMSCs can modulate the phenotype and angiogenic function of macrophages. In vitro, cMSCs induce apoptosis of macrophages while preferentially promoting a distinct CD14hi CD16hi CD163hi CD206hi immunophenotype that has significantly reduced angiogenic effects based on in vitro angiogenesis assays. In vivo, application of cMSCs to murine corneas after injury leads to reduced macrophage infiltration and higher expression of CD206 in macrophages. Macrophages cocultured ("educated") by cMSCs express significantly higher levels of anti-angiogenic and anti-inflammatory factors compared with control macrophages. In vivo, injured corneas treated with cMSC-educated macrophages demonstrate significantly less neovascularization compared with corneas treated with control macrophages. Knocking down the expression of pigment epithelial derived factor (PEDF) in cMSCs significantly abrogates its modulating effects on macrophages, as shown by the reduced rate of apoptosis, decreased expression of sFLT-1/PEDF, and increased expression of vascular endothelial growth factor-A in the cocultured macrophages. Similarly, cMSCs isolated from PEDF knockout mice are less effective compared with wild-type cMSCs at inhibiting macrophage infiltration when applied to wild-type corneas after injury. Overall, these results demonstrate that cMSCs therapeutically suppress the angiogenic capacity of macrophages and highlight the role of cMSC secreted PEDF in the modulation of macrophage phenotype and function. Stem Cells 2018;36:775-784.


Subject(s)
Cornea/cytology , Immunomodulation/physiology , Macrophages/cytology , Mesenchymal Stem Cells/cytology , Neovascularization, Physiologic , Animals , Apoptosis/physiology , Cornea/blood supply , Immunophenotyping/methods , Mice, Knockout
11.
Invest Ophthalmol Vis Sci ; 58(12): 5507-5517, 2017 10 01.
Article in English | MEDLINE | ID: mdl-29075761

ABSTRACT

Purpose: To evaluate the angiogenic properties of corneal derived mesenchymal stromal cells (Co-MSC). Methods: Co-MSCs were extracted from human cadaver, and wild-type (C57BL/6J) and SERPINF1-/- mice corneas. The MSC secretome was collected in a serum-free medium. Human umbilical vein endothelial cell (HUVEC) tube formation and fibrin gel bead assay (FIBA) sprout formation were used to assess the angiogenic properties of Co-MSC secretome. Complete corneal epithelial debridement was used to induce corneal neovascularization in wild-type mice. Co-MSCs embedded in fibrin gel was applied over the debrided cornea to evaluate the angiogenic effects of Co-MSCs in vivo. Immunoprecipitation was used to remove soluble fms-like tyrosine kinase-1 (sFLT-1) and pigment epithelium-derived factor (PEDF, SERPINF1 gene) from the Co-MSC secretome. Results: Co-MSC secretome significantly inhibited HUVECs tube and sprout formation. Co-MSCs from different donors consistently contained high levels of antiangiogenic factors including sFLT-1 and PEDF; and low levels of the angiogenic factor VEGF-A. In vivo, application of Co-MSCs to mouse corneas after injury prevented the development of corneal neovascularization. Removing PEDF or sFLT-1 from the secretome significantly diminished the antiangiogenic effects of Co-MSCs. Co-MSCs isolated from SERPINF1-/- mice had significantly reduced antiangiogenic effects compared to SERPINF1+/+ (wild-type) Co-MSCs. Conclusions: These results illustrate the direct antiangiogenic properties of Co-MSCs, the importance of sFLT-1 and PEDF, and their potential clinical application for preventing pathologic corneal neovascularization.


Subject(s)
Cornea/metabolism , Corneal Neovascularization/metabolism , Eye Proteins/biosynthesis , Mesenchymal Stem Cells/metabolism , Nerve Growth Factors/biosynthesis , Serpins/biosynthesis , Vascular Endothelial Growth Factor Receptor-1/biosynthesis , Animals , Blotting, Western , Cadaver , Cells, Cultured , Cornea/pathology , Corneal Neovascularization/pathology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Immunoprecipitation , Mesenchymal Stem Cells/cytology , Mice, Inbred C57BL
12.
ACS Appl Mater Interfaces ; 9(28): 23389-23399, 2017 Jul 19.
Article in English | MEDLINE | ID: mdl-28598594

ABSTRACT

Surface modifications with tethered growth factors have mainly been applied to synthetic polymeric biomaterials in well-controlled, acellular settings, followed by seeding with cells. The known bio-orthogonality of copper-free click chemistry provides an opportunity to not only use it in vitro to create scaffolds or pro-migratory tracks in the presence of living cells, but also potentially apply it to living tissues directly as a coupling modality in situ. In this study, we studied the chemical coupling of growth factors to collagen using biocompatible copper-free click chemistry and its effect on the enhancement of growth factor activity in vitro. We verified the characteristics of modified epidermal growth factor (EGF) using mass spectrometry and an EGF/EGF receptor binding assay, and evaluated the chemical immobilization of EGF on collagen by copper-free click chemistry using surface X-ray photoelectron spectroscopy (XPS), surface plasmon resonance (SPR) spectroscopy, and enzyme-linked immunosorbent assay (ELISA). We found that the anchoring was noncytotoxic, biocompatible, and rapid. Moreover, the surface-immobilized EGF had significant effects on epithelial cell attachment and proliferation. Our results demonstrate the possibility of copper-free click chemistry as a tool for covalent bonding of growth factors to collagen in the presence of living cells. This approach is a novel and potentially clinically useful application of copper-free click chemistry as a way of anchoring growth factors to collagen and foster epithelial wound healing.


Subject(s)
Collagen/chemistry , Alkynes , Azides , Biocompatible Materials , Click Chemistry , Mass Spectrometry
13.
Sci Rep ; 7: 40308, 2017 01 05.
Article in English | MEDLINE | ID: mdl-28054657

ABSTRACT

Rapamycin has previously been shown to have anti-aging effects in cells and organisms. These studies were undertaken to investigate the effects of rapamycin on primary human corneal epithelial cells in vitro. Cell growth and viability were evaluated by bright field microscopy. Cell proliferation and cycle were evaluated by flow cytometry. The expression of differentiation markers was evaluated by quantitative PCR and Western blot. Senescence was evaluated by senescence-associated ß-Galactosidase staining and by Western blot analysis of p16. Apoptosis was evaluated by a TUNEL assay. The results demonstrated that primary HCEC treated with rapamycin had lower proliferation but considerably longer survival in vitro. Rapamycin-treated cells maintained a higher capacity to proliferate after removal of rapamycin and expressed more keratin 14, N-Cadherin, DeltaNp63 and ABCG2, and less keratin 12, consistent with their less differentiated state. Rapamycin treated cells demonstrated less senescence by X-ß-Gal SA staining and by lower expression of p16. Apoptosis was also lower in the rapamycin treated cells. These results indicate that rapamycin treatment of HCEC prevents the loss of corneal epithelial stem/progenitor cells to replicative senescence and apoptosis. Rapamycin may be a useful additive for ex vivo expansion of corneal epithelial cells.


Subject(s)
Epithelial Cells/cytology , Epithelium, Corneal/cytology , Sirolimus/pharmacology , Adolescent , Adult , Aged , Aged, 80 and over , Apoptosis/drug effects , Biomarkers/metabolism , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Cellular Senescence/drug effects , Epithelial Cells/drug effects , Humans , Inflammation/pathology , Limbus Corneae/cytology , Middle Aged , Young Adult
14.
Biochim Biophys Acta ; 1860(10): 2148-56, 2016 10.
Article in English | MEDLINE | ID: mdl-27233452

ABSTRACT

BACKGROUND: Immunohistochemical staining experiments have shown that both hemangiogenesis and lymphangiogenesis occur following severe corneal and conjunctival injury and that the neovascularization of the cornea often has severe visual consequences. To better understand how hemangiogenesis and lymphangiogenesis are induced by different degrees of ocular injury, we investigated patterns of injury-induced corneal neovascularization in live Prox1-GFP/Flk1::myr-mCherry mice, in which blood and lymphatic vessels can be imaged simultaneously in vivo. METHODS: The eyes of Prox1-GFP/Flk1::myr-mCherry mice were injured according to four models based on epithelial debridement of the: A) central cornea (a 1.5-mm-diameter circle of tissue over the corneal apex), B) total cornea, C) bulbar conjunctiva, and D) cornea+bulbar conjunctiva. Corneal blood and lymphatic vessels were imaged on days 0, 3, 7, and 10 post-injury, and the percentages of the cornea containing blood and lymphatic vessels were calculated. RESULTS: Neither central corneal nor bulbar conjunctival debridement resulted in significant vessel growth in the mouse cornea, whereas total corneal and corneal+bulbar conjunctival debridement did. On day 10 in the central cornea, total cornea, bulbar conjunctiva, and corneal+bulbar conjunctival epithelial debridement models, the percentage of the corneal surface that was occupied by blood vessels (hemangiogenesis) was 1.9±0.8%, 7.14±2.4%, 2.29±1%, and 15.05±2.14%, respectively, and the percentage of the corneal surface that was occupied by lymphatic vessels (lymphangiogenesis) was 2.45±1.51%, 4.85±0.95%, 2.95±1.27%, and 4.15±3.85%, respectively. CONCLUSIONS: Substantial corneal debridement was required to induce corneal neovascularization in the mouse cornea, and the corneal epithelium may therefore be partially responsible for maintaining corneal avascularity. GENERAL SIGNIFICANCE: Our study demonstrates that GFP/Flk1::myr-mCherry mice are a useful model for studying coordinated hemangiogenic and lymphangiogenic responses.


Subject(s)
Cornea/growth & development , Corneal Injuries/genetics , Corneal Neovascularization/genetics , Homeodomain Proteins/genetics , Tumor Suppressor Proteins/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics , Animals , Cornea/metabolism , Cornea/pathology , Corneal Injuries/pathology , Corneal Neovascularization/pathology , Debridement , Disease Models, Animal , Epithelium, Corneal/growth & development , Epithelium, Corneal/pathology , Humans , Lymphangiogenesis/genetics , Lymphatic Vessels/metabolism , Lymphatic Vessels/pathology , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...