Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-38899506

ABSTRACT

Purpose: The present study aimed to determine the dose-response relationship between targeted nanocarriers released from a novel, sustained release formulation and their ability to specifically deplete cells responsible for the development of posterior capsular opacification (PCO) in month-long, dynamic cell cultures. Methods: Injectable, thermosensitive poly(D,L-lactic-co-glycolic acid)-b-poly(ethylene glycol)-b-poly(D,L-lactic-co-glycolic acid) triblock copolymer hydrogels were loaded with either a low or a high dose of doxorubicin-loaded antibody-targeted nanocarriers (G8:3DNA:Dox). Human rhabdomyosarcoma cells, selected for their expression of PCO marker brain-specific angiogenesis inhibitor 1 (BAI1), were kept under dynamic media flow and received either a low or high dose of nanocarriers. Cells were fixed and stained at predetermined time points to evaluate targeted depletion of BAI1+ cells. Results: A lower dose of nanocarriers in hydrogel depleted BAI1+ cells at a slower rate than the higher dose, whereas both reached over 90% BAI1+ cellular nonviability at 28 days. Both treatment groups also significantly lowered the relative abundance of BAI1+ cells in the population compared with the control group. Conclusions: Controlled release of a lower dose of nanocarriers can still achieve therapeutically relevant effects in the prevention of PCO, while avoiding potential secondary effects associated with the administration of a higher dose.

2.
Cells ; 12(13)2023 06 27.
Article in English | MEDLINE | ID: mdl-37443759

ABSTRACT

Herein, we review a unique and versatile lineage composed of Myo/Nog cells that may be beneficial or detrimental depending on their environment and nature of the pathological stimuli they are exposed to. While we will focus on the lens, related Myo/Nog cell behaviors and functions in other tissues are integrated into the narrative of our research that spans over three decades, examines multiple species and progresses from early stages of embryonic development to aging adults. Myo/Nog cells were discovered in the embryonic epiblast by their co-expression of the skeletal muscle-specific transcription factor MyoD, the bone morphogenetic protein inhibitor Noggin and brain-specific angiogenesis inhibitor 1. They were tracked from the epiblast into the developing lens, revealing heterogeneity of cell types within this structure. Depletion of Myo/Nog cells in the epiblast results in eye malformations arising from the absence of Noggin. In the adult lens, Myo/Nog cells are the source of myofibroblasts whose contractions produce wrinkles in the capsule. Eliminating this population within the rabbit lens during cataract surgery reduces posterior capsule opacification to below clinically significant levels. Parallels are drawn between the therapeutic potential of targeting Myo/Nog cells to prevent fibrotic disease in the lens and other ocular tissues.


Subject(s)
Capsule Opacification , Lens, Crystalline , Animals , Rabbits , Lens, Crystalline/metabolism , Capsule Opacification/metabolism , Muscle, Skeletal/metabolism , Receptors, G-Protein-Coupled/metabolism , Gene Expression Regulation
3.
Invest Ophthalmol Vis Sci ; 64(2): 1, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36723927

ABSTRACT

Purpose: Myo/Nog cells are the source of myofibroblasts in the lens and synthesize muscle proteins in human epiretinal membranes (ERMs). In the current study, we examined the response of Myo/Nog cells during ERM formation in a mouse model of proliferative vitreoretinopathy (PVR). Methods: PVR was induced by intravitreal injections of gas and ARPE-19 cells. PVR grade was scored by fundus imaging, optical coherence tomography, and histology. Double label immunofluorescence localization was performed to quantify Myo/Nog cells, myofibroblasts, and leukocytes. Results: Myo/Nog cells, identified by co-labeling with antibodies to brain-specific angiogenesis inhibitor 1 (BAI1) and Noggin, increased throughout the eye with induction of PVR and disease progression. They were present on the inner surface of the retina in grades 1/2 PVR and were the largest subpopulation of cells in grades 3 to 6 ERMs. All α-SMA-positive (+) cells and all but one striated myosin+ cell expressed BAI1 in grades 1 to 6 PVR. Folds and areas of retinal detachment were overlain by Myo/Nog cells containing muscle proteins. Low numbers of CD18, CD68, and CD45+ leukocytes were detected throughout the eye. Small subpopulations of BAI1+ cells expressed leukocyte markers. ARPE-19 cells were found in the vitreous but were rare in ERMs. Pigmented cells lacking Myo/Nog and muscle cell markers were present in ERMs and abundant within the retina by grade 5/6. Conclusions: Myo/Nog cells differentiate into myofibroblasts that appear to contract and produce retinal folds and detachment. Targeting BAI1 for Myo/Nog cell depletion may be a pharmacological approach to preventing and treating PVR.


Subject(s)
Epiretinal Membrane , Vitreoretinopathy, Proliferative , Animals , Mice , Humans , Vitreoretinopathy, Proliferative/metabolism , Epiretinal Membrane/metabolism , Myofibroblasts/metabolism , Retina/metabolism , Muscle Proteins/metabolism
4.
J Ocul Pharmacol Ther ; 38(6): 404-411, 2022.
Article in English | MEDLINE | ID: mdl-35377237

ABSTRACT

Purpose: To compare a novel, sustained release formulation and a bolus injection of a targeted nanocarrier for the ability to specifically deplete cells responsible for the development of posterior capsule opacification (PCO) in week-long, dynamic cell cultures. Methods: A novel, injectable, thermosensitive poly(D,L-lactic-co-glycolic acid)-b-poly(ethylene glycol)-b-poly(D,L-lactic-co-glycolic acid) (PLGA-PEG-PLGA) triblock copolymer hydrogel was engineered for the sustained release of targeted, nucleic acid nanocarriers loaded with cytotoxic doxorubicin (G8:3DNA:Dox). Human rhabdomyosarcoma (RD) cells were used due to their expression of brain-specific angiogenesis inhibitor 1 (BAI1), a specific marker for the myofibroblasts responsible for PCO. Under constant media flow, nanocarriers were injected into cell cultures as either a bolus or within the hydrogel. Cells were fixed and stained every other day for 7 days to compare targeted depletion of BAI1+ cells. Results: The formulation transitions to a gel at physiological temperatures, is optically clear, noncytotoxic, and can release G8:3DNA:Dox nanocarriers for up to 4 weeks. In RD cell cultures, G8:3DNA:Dox nanocarriers specifically eliminated BAI1+ cells. The bolus nanocarrier dose showed significantly reduced cell depletion overtime, while the sustained release of nanocarriers showed increased cell depletion over time. By day 7, <2% of BAI1+ cells were depleted by the bolus injection and 74.2% BAI1+ cells were targeted by the sustained release of nanocarriers. Conclusions: The sustained release of nanocarriers from the hydrogel allows for improved therapeutic delivery in a dynamic system. This method can offer a more effective and efficient method of prophylactically treating PCO after cataract surgery.


Subject(s)
Capsule Opacification , Hydrogels , DNA , Delayed-Action Preparations , Doxorubicin , Humans , Lactic Acid , Polyethylene Glycols
5.
Front Neurosci ; 15: 780707, 2021.
Article in English | MEDLINE | ID: mdl-34949984

ABSTRACT

Focal brain injury in the form of a needlestick (NS) results in cell death and induces a self-protective response flanking the lesion. Myo/Nog cells are identified by their expression of bone morphogenetic protein inhibitor Noggin, brain-specific angiogenesis inhibitor 1 (BAI1) and the skeletal muscle specific transcription factor MyoD. Myo/Nog cells limit cell death in two forms of retinopathy. In this study, we examined the acute response of Myo/Nog cells to a NS lesion that extended from the rat posterior parietal cortex to the hippocampus. Myo/Nog cells were identified with antibodies to Noggin and BAI1. These cells were the primary source of both molecules in the uninjured and injured brain. One day after the NS, the normally small population of Myo/Nog cells expanded approximately eightfold within a 1 mm area surrounding the lesion. Myo/Nog cells were reduced by approximately 50% along the lesion with an injection of the BAI1 monoclonal antibody and complement. The number of dying cells, identified by terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling (TUNEL), was unchanged at this early time point in response to the decrease in Myo/Nog cells. However, increasing the number of Myo/Nog cells within the lesion by injecting BAI1-positive (+) cells isolated from the brains of other animals, significantly reduced cell death and increased the number of NeuN+ neurons compared to brains injected with phosphate buffered saline or exogenous BAI1-negative cells. These findings demonstrate that Myo/Nog cells rapidly react to injury within the brain and increasing their number within the lesion is neuroprotective.

6.
PLoS One ; 15(8): e0235898, 2020.
Article in English | MEDLINE | ID: mdl-32833999

ABSTRACT

Myo/Nog cells were discovered in the chick embryo epiblast. Their expression of MyoD reflects a commitment to the skeletal muscle lineage and capacity to differentiate into myofibroblasts. Release of Noggin by Myo/Nog cells is essential for normal morphogenesis. Myo/Nog cells rapidly respond to wounding in the skin and eyes. In this report, we present evidence suggesting that Myo/Nog cells phagocytose tattoo ink in tissue sections of human skin and engulf cell corpses in cultures of anterior human lens tissue and magnetic beads injected into the anterior chamber of mice in vivo. Myo/Nog cells are distinct from macrophages in the skin and eyes indicated by the absence of labeling with an antibody to ionized calcium binding adaptor molecule 1. In addition to their primary roles as regulators of BMP signaling and progenitors of myofibroblasts, Myo/Nog cells behave as nonprofessional phagocytes defined as cells whose primary functions are unrelated to phagocytosis but are capable of engulfment.


Subject(s)
Myofibroblasts/cytology , Phagocytes/cytology , Stem Cells/cytology , Animals , Carrier Proteins/metabolism , Cell Differentiation , Cells, Cultured , Chick Embryo , Female , Humans , Lens, Crystalline/cytology , Lens, Crystalline/metabolism , Male , Mice , Mice, Inbred C57BL , MyoD Protein/metabolism , Myofibroblasts/metabolism , Phagocytes/metabolism , Phagocytosis , Rabbits , Skin/cytology , Skin/metabolism , Stem Cells/metabolism
7.
PLoS One ; 15(7): e0234792, 2020.
Article in English | MEDLINE | ID: mdl-32614850

ABSTRACT

The Myo/Nog cell lineage was discovered in the chick embryo and is also present in adult mammalian tissues. The cells are named for their expression of mRNA for the skeletal muscle specific transcription factor MyoD and bone morphogenetic protein inhibitor Noggin. A third marker for Myo/Nog cells is the cell surface molecule recognized by the G8 monoclonal antibody (mAb). G8 has been used to detect, track, isolate and kill Myo/Nog cells. In this study, we screened a membrane proteome array for the target of the G8 mAb. The array consisted of >5,000 molecules, each synthesized in their native confirmation with appropriate post-translational modifications in a single clone of HEK-293T cells. G8 mAb binding to the clone expressing brain-specific angiogenesis inhibitor 1 (BAI1) was detected by flow cytometry, re-verified by sequencing and validated by transfection with the plasmid construct for BAI1. Further validation of the G8 target was provided by enzyme-linked immunosorbent assay. The G8 epitope was identified by screening a high-throughput, site directed mutagenesis library designed to cover 95-100% of the 954 amino acids of the extracellular domain of the BAI1 protein. The G8 mAb binds within the third thrombospondin repeat of the extracellular domain of human BAI1. Immunofluorescence localization experiments revealed that G8 and a commercially available BAI1 mAb co-localize to the subpopulation of Myo/Nog cells in the skin, eyes and brain. Expression of the multi-functional BAI1 protein in Myo/Nog cells introduces new possibilities for the roles of Myo/Nog cells in normal and diseased tissues.


Subject(s)
Angiogenic Proteins/biosynthesis , Myofibroblasts/metabolism , Receptors, G-Protein-Coupled/biosynthesis , Amino Acid Substitution , Angiogenic Proteins/chemistry , Angiogenic Proteins/genetics , Angiogenic Proteins/immunology , Animals , Antibodies, Monoclonal/immunology , Antibody Specificity , Antigen-Antibody Reactions , Brain/cytology , Carrier Proteins/analysis , Cell Lineage , Epitopes/immunology , Eye Proteins/biosynthesis , Eye Proteins/chemistry , Eye Proteins/genetics , Eye Proteins/immunology , Humans , Male , Mice , Mice, Inbred C57BL , Models, Molecular , Muscle Development , MyoD Protein/analysis , Organ Specificity , Protein Conformation , Protein Domains , Rabbits , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/immunology , Repetitive Sequences, Amino Acid , Skin/cytology , Species Specificity , Tattooing , Young Adult
8.
Exp Eye Res ; 197: 108080, 2020 08.
Article in English | MEDLINE | ID: mdl-32474138

ABSTRACT

Proliferative vitreoretinopathy (PVR) is a complication of rhegmatogenous retinal detachment and ocular trauma. The disease is characterized by development of membranes that may apply traction to the retina and cause redetachment. Membrane contractions are attributed to myofibroblasts arising from retinal pigment epithelial cells, glia and fibroblasts. The progenitors of myofibrobasts in the lens are Myo/Nog cells that express the skeletal muscle transcription factor MyoD and bone morphogenetic protein inhibitor Noggin. The retina and choroid also contain Myo/Nog cells that respond to stress. We examined preretinal PVR membranes from three ocular trauma patients with retinal detachment for Myo/Nog cells and their expression of muscle proteins. Myo/Nog cells were identified by co-localization of antibodies to the G8 antigen and Noggin. Greater than 80% of all cells in sections from two of three patients expressed both G8 and Noggin. Myo/Nog cells lacked pigment. Alpha smooth muscle actin (α-SMA) and striated myosin II heavy chain were present in the majority of Myo/Nog cells in these two patients. Differentiation of Myo/Nog cells was paralleled by low levels of MyoD. Membrane sections from the third patient consisted mostly of connective tissue with very few cells. A small subpopulation in these sections expressed both G8 and Noggin, and muscle proteins were detected in only a minority of G8-positive (+) cells. In all three patients, greater than 99% of cells with MyoD, α-SMA and striated muscle myosin co-expressed G8. These findings suggest that contractile myofibroblasts in PVR membranes may be derived from differentiating Myo/Nog cells.


Subject(s)
Muscle Proteins/biosynthesis , MyoD Protein/biosynthesis , Retina/pathology , Vitreoretinopathy, Proliferative/metabolism , Animals , Humans , Retina/metabolism , Vitreoretinopathy, Proliferative/diagnosis
11.
J Am Osteopath Assoc ; 119(9): 588-596, 2019 Sep 01.
Article in English | MEDLINE | ID: mdl-31449305

ABSTRACT

CONTEXT: Analyzing factors that may enhance osteopathic applicants' likelihood of matching is warranted given that United States osteopathic and allopathic residency programs will have a single accreditation system in 2020. OBJECTIVES: To determine the impact of research accomplishments and experiences on osteopathic and allopathic residency matching. METHODS: Analysis of variance, t test, and odds ratios were used to examine data from the National Resident Matching Program Charting Outcomes from 2016 and 2018. Relationships between match status and medical degree, specialty matching, and mean numbers of research accomplishments and experiences in the Main Residency Match were analyzed. RESULTS: Matched osteopathic and allopathic applicants had significantly greater numbers of research accomplishments (mean [SD], 5.18 [4.34]) than unmatched applicants (3.66 [2.87]) (P=.006). Applicants who matched (mean [SD], 2.81 [1.64]) had similar numbers of research experiences to those who did not match (2.43 [1.26]) (P=.068). Matched and unmatched allopathic applicants' research accomplishments (5.91 [3.72]) were significantly greater than that of osteopathic applicants (2.60 [2.90]) (P<.001). Significant differences also were found between the means of research experiences of matched and unmatched osteopathic (mean [SD], 1.73 [1.21]) and allopathic applicants (3.36 [1.25], P<.001). Matched and unmatched osteopathic applicants' had similar means for research accomplishments (mean [SD], 3.00 [3.64] and 2.20 [1.84], respectively; P=.242) and experiences (1.79 [1.31] and 1.66 [1.12], respectively; P=.664). By contrast, significant differences were found between the numbers of research accomplishments for matched (mean [SD], 6.97 [4.07]) vs unmatched (4.86 [3.02]) allopathic applicants (P=.007). The only subspecialty for which research experiences of osteopathic applicants correlated with matching was physical medicine and rehabilitation (OR, 2.75; 95% CI, 1.30-5.84). CONCLUSION: Research seems to have a greater influence on matching for allopathic than osteopathic applicants. Although both osteopathic and allopathic programs have standards pertaining to scholarly activity, allopathic medical schools may place a greater emphasis on research. Increasing osteopathic medical students' exposure to research is predicted to enhance their competitiveness for matching and help develop skills relevant to the practice of evidence-based medicine.


Subject(s)
Career Choice , Internship and Residency , Osteopathic Medicine/education , Personnel Selection , Research , School Admission Criteria , Accreditation , Education, Medical , Humans , United States
12.
J Am Osteopath Assoc ; 119(9): 598-611, 2019 Sep 01.
Article in English | MEDLINE | ID: mdl-31449306

ABSTRACT

The osteopathic undergraduate medical education standards have evolved over the past 2 decades to require undergraduate medical student participation in research and scholarly activity. The authors' objective was to review those evolving standards and develop a model for introducing the principles and practice of research that combines core content with experiential learning. They identified fundamental topics pertinent to the research process and herein provide their recommendations for incorporating these topics into the curriculum as self-study, online modules, and team-based and active learning exercises. The authors' proposed educational model would provide an avenue for osteopathic medical schools to meet the requirements for scholarly activity. Increasing students' and residents' knowledge of the research process will lay the foundation for their engagement is research and scholarly activity and their practice of evidence-based medicine.


Subject(s)
Education, Medical, Undergraduate/standards , Models, Educational , Osteopathic Medicine/standards , Research , Humans , Program Development
13.
Invest Ophthalmol Vis Sci ; 60(6): 1813-1823, 2019 05 01.
Article in English | MEDLINE | ID: mdl-31042787

ABSTRACT

Purpose: Posterior capsule opacification (PCO) is a vision-impairing disease that occurs in some adults and most children after cataract surgery. Contractile myofibroblasts contribute to PCO by producing wrinkles in the lens capsule that scatter light. Myofibroblasts in the lens originate from Myo/Nog cells named for their expression of the MyoD transcription factor and bone morphogenetic protein inhibitor noggin. In this study we tested the effects of depleting Myo/Nog cells on development of PCO. Methods: Myo/Nog cells were eliminated by injecting the G8 antibody conjugated to 3DNA nanocarriers for the cytotoxin doxorubicin (G8:3DNA:Dox) during cataract surgery in rabbits. The severity of PCO was scored by slit lamp analysis, gross and histologic observation, and immunofluorescence localization of α-smooth muscle actin. Results: G8:3DNA:Dox specifically induced cell death in Myo/Nog cells in the lens. None of the lenses administered G8:3DNA containing 9 to 36 µM doxorubicin developed greater than trace levels of central PCO and few myofibroblasts were present on the capsule. Less than 9% of these lenses exhibited greater than mild levels of peripheral PCO. Doxorubucin itself reduced PCO; however, myofibroblasts and wrinkles were abundant in the lens, and off-target effects were observed in the ciliary processes and cornea. Conclusions: Myo/Nog cells are the primary source of myofibroblasts in the lens after cataract surgery. Targeted depletion of Myo/Nog cells has potential for preventing PCO and preserving vision.


Subject(s)
Capsule Opacification/pathology , Carrier Proteins/metabolism , MyoD Protein/metabolism , Myofibroblasts/pathology , Posterior Capsule of the Lens/pathology , Animals , Capsule Opacification/metabolism , Disease Models, Animal , Female , Myofibroblasts/metabolism , Posterior Capsule of the Lens/metabolism , Rabbits
14.
PLoS One ; 14(4): e0214758, 2019.
Article in English | MEDLINE | ID: mdl-30973903

ABSTRACT

Myo/Nog cells are identified by their expression of the skeletal muscle specific transcription factor MyoD and the bone morphogenetic protein inhibitor noggin, and binding of the G8 monoclonal antibody. Their release of noggin is critical for morphogenesis and skeletal myogenesis. In the adult, Myo/Nog cells are present in normal tissues, wounds and skin tumors. Myo/Nog cells in the lens give rise to myofibroblasts that synthesize skeletal muscle proteins. The purpose of this study was to screen human lens tissue, rhabdomyosarcoma cell lines, and tissue sections from rhabdomyosarcoma, Wilms and tumors lacking features of skeletal muscle for co-localization of antibodies to Myo/Nog cell markers and the lens beaded filament proteins filensin and CP49. Immunofluorescence localization experiments revealed that Myo/Nog cells of the lens bind antibodies to beaded filament proteins. Co-localization of antibodies to G8, noggin, filensin and CP49 was observed in most RC13 and a subpopulation of RD human rhabdomyosarcoma cell lines. Western blotting with beaded filament antibodies revealed bands of similar molecular weights in RC13 and murine lens cells. Human alveolar, embryonal, pleomorphic and spindle cell rhabdomyosarcomas and Wilms tumors contained a subpopulation of cells immunoreactive for G8, noggin, MyoD and beaded filaments. G8 was also co-localized with filensin mRNA. Staining for beaded filament proteins was not detected in G8 positive cells in leiomyosarcomas, squamous and basal cell carcinomas, syringocarciomas and malignant melanomas. Lens beaded filament proteins were thought to be present only in the lens. Myo/Nog-like cells immunoreactive for beaded filaments may be diagnostic of tumors related to the skeletal muscle lineage.


Subject(s)
Carrier Proteins/metabolism , Eye Proteins/metabolism , Intermediate Filament Proteins/metabolism , MyoD Protein/metabolism , Rhabdomyosarcoma/pathology , Wilms Tumor/pathology , Animals , Antibodies, Monoclonal/immunology , Carrier Proteins/immunology , Cell Line , Eye Proteins/genetics , Eye Proteins/immunology , Humans , Intermediate Filament Proteins/genetics , Intermediate Filament Proteins/immunology , Lens, Crystalline/cytology , Lens, Crystalline/metabolism , Mice , Microscopy, Fluorescence , MyoD Protein/immunology , Rhabdomyosarcoma/metabolism , Rhabdomyosarcoma, Embryonal/metabolism , Rhabdomyosarcoma, Embryonal/pathology , Wilms Tumor/metabolism
15.
Exp Eye Res ; 171: 101-105, 2018 06.
Article in English | MEDLINE | ID: mdl-29559302

ABSTRACT

Myo/Nog cells, named for their expression of MyoD and noggin, enter the eye during early stages of embryonic development. Their release of noggin is critical for normal morphogenesis of the lens and retina. Myo/Nog cells are also present in adult eyes. Single nucleated skeletal muscle cells designated as myofibroblasts arise from Myo/Nog cells in cultures of lens tissue. In this report we document the presence of Myo/Nog cells in the lens, ciliary body and on the zonule of Zinn in mice, rabbits and humans. Myo/Nog cells were rare in all three structures. Their prevalence increased in the lens and ciliary body of rabbits 24 h following cataract surgery. Rabbits developed posterior capsule opacification (PCO) within one month of surgery. The number of Myo/Nog cells continued to be elevated in the lens and ciliary body. Myo/Nog cells containing alpha smooth muscle actin and striated muscle myosin were present on the posterior capsule and overlaid deformations in the capsule. Myo/Nog cells also were present on the zonule fibers and external surface of the posterior capsule. These findings suggest that Myo/Nog contribute to PCO and may use the zonule fibers to migrate between the ciliary processes and lens.


Subject(s)
Carrier Proteins/metabolism , Ciliary Body/metabolism , Lens, Crystalline/metabolism , Ligaments/metabolism , MyoD Protein/metabolism , Phacoemulsification , Posterior Capsule of the Lens/metabolism , Actins/metabolism , Animals , Capsule Opacification/metabolism , Female , Fibrillin-1/metabolism , Fluorescent Antibody Technique, Indirect , Humans , Mice , Mice, Inbred C57BL , Myofibroblasts/metabolism , Myosins/metabolism , Rabbits , Vimentin/metabolism
16.
Dose Response ; 16(4): 1559325818803428, 2018.
Article in English | MEDLINE | ID: mdl-30627064

ABSTRACT

This review brings together observations on the stress-induced regulation of resilience mechanisms in body tissues. It is argued that the stresses that induce tissue resilience in mammals arise from everyday sources: sunlight, food, lack of food, hypoxia and physical stresses. At low levels, these stresses induce an organised protective response in probably all tissues; and, at some higher level, cause tissue destruction. This pattern of response to stress is well known to toxicologists, who have termed it hormesis. The phenotypes of resilience are diverse and reports of stress-induced resilience are to be found in journals of neuroscience, sports medicine, cancer, healthy ageing, dementia, parkinsonism, ophthalmology and more. This diversity makes the proposing of a general concept of induced resilience a significant task, which this review attempts. We suggest that a system of stress-induced tissue resilience has evolved to enhance the survival of animals. By analogy with acquired immunity, we term this system 'acquired resilience'. Evidence is reviewed that acquired resilience, like acquired immunity, fades with age. This fading is, we suggest, a major component of ageing. Understanding of acquired resilience may, we argue, open pathways for the maintenance of good health in the later decades of human life.

17.
Exp Cell Res ; 357(2): 310-319, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28583763

ABSTRACT

Osteoarthritis (OA) is characterized by degeneration of articular cartilage within the joint, inflammation and pain. The purpose of this study was to develop a primary, serum free cell culture system of human osteoarthritic articular chondrocytes (HOACs) with which to study manifestations of the disease process. Joint tissues were obtained from OA patients undergoing total knee arthroplasty (TKA). HOACs isolated from the femoral condyles and tibial plateau of the same side were combined, plated in three-dimensional, alginate beads and cultured for five days in serum, hormone and protein free medium. More living cells were obtained from the femoral condyles than the tibial plateau. The optimal plating density was 2.5 × 106 cells/ml of alginate. The amounts of DNA, RNA, proteoglycans and total collagen were similar in cultures prepared from the sides of least and greatest pathology. More type 1 than type 2 collagen was detected in the medium on days 2 and 5. A greater percentage of type 1 than type 2 collagen was degraded. The inflammatory cytokine interleukin-1 beta was present in the medium and alginate associated matrix. Although variation in the metabolic profiles between subjects was observed, HOACs from all patients continued to reflect the OA phenotype for five days in culture. This serum free, three-dimensional primary culture system of HOACs provides a platform with which to measure clinically relevant endpoints of OA and screen potential disease modifying OA therapeutics.


Subject(s)
Cartilage, Articular/cytology , Chondrocytes/metabolism , Osteoarthritis/metabolism , Primary Cell Culture , Proteoglycans/metabolism , Collagen/metabolism , Collagen Type II/metabolism , Culture Media, Serum-Free , Extracellular Matrix/metabolism , Humans
18.
J Pharmacol Exp Ther ; 361(1): 60-67, 2017 04.
Article in English | MEDLINE | ID: mdl-28096456

ABSTRACT

Posterior capsule opacification (PCO) occurs in some adults and most children following cataract surgery. The fibrotic form of PCO arises, in part, from migratory, contractile myofibroblasts that deform the lens capsule and impair vision. In short-term cultures of human anterior lens tissue, myofibroblasts emerge from Myo/Nog cells that are identified with the G8 monoclonal antibody and by their expression of the MyoD transcription factor and bone morphogenetic protein inhibitor noggin. In this study, we tested the hypothesis that targeted depletion of Myo/Nog cells with the G8 monoclonal antibody (mAb) conjugated to three-dimensional DNA nanocarriers intercalated with doxorubicin (G8:3DNA:Dox) would prevent the accumulation of myofibroblasts in long-term, serum- and growth factor-free cultures of human lens tissue obtained by capsulorhexis. The mAb:nanocarrier complex was internalized into acidic compartments of the cell. G8:3DNA:Dox killed nearly all Myo/Nog cells without affecting the lens epithelial cells. In 30-day cultures, all G8-positive cells expressed noggin, and subpopulations had synthesized MyoD, sarcomeric myosin, and alpha smooth muscle actin (α-SMA). Myo/Nog cells responded to scratching of the lens epithelium by accumulating around the edges of the wound. Treatment with two doses of G8:3DNA:Dox completely eliminated G8+/α-SMA+ cells throughout the explant. These experiments demonstrate that Myo/Nog cells are the source of myofibroblasts in long-term cultures of anterior human lens tissue and mAb:3DNA nanocarriers specifically and effectively deliver cytotoxic cargo to a subpopulation of cells without off-target effects. G8:3DNA:Dox has the potential to reduce PCO following cataract surgery.


Subject(s)
Antibodies, Monoclonal/administration & dosage , DNA/administration & dosage , Doxorubicin/administration & dosage , Lens, Crystalline/drug effects , Myofibroblasts/drug effects , Nanostructures/administration & dosage , Aged , Aged, 80 and over , Antibodies, Monoclonal/metabolism , DNA/metabolism , Doxorubicin/metabolism , Drug Carriers/administration & dosage , Drug Carriers/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , Humans , Lens, Crystalline/cytology , Lens, Crystalline/metabolism , Male , Middle Aged , Myofibroblasts/metabolism , Organ Culture Techniques
19.
PLoS One ; 12(1): e0169744, 2017.
Article in English | MEDLINE | ID: mdl-28099524

ABSTRACT

PURPOSE: To identify Myo/Nog cells in the adult retina and test their role in protecting retinal photoreceptors from light damage. METHODS: Light damage was induced by exposing albino rats raised in dim cyclic light to 1000 lux light for 24 hours. In one group of rats, Myo/Nog cells were purified from rat brain tissue by magnetic cell sorting following binding of the G8 monoclonal antibody (mAb). These cells were injected into the vitreous humour of the eye within 2 hours following bright light exposure. Retinal function was assessed using full-field, flash electroretinogram (ERG) before and after treatment. The numbers of Myo/Nog cells, apoptotic photoreceptors, and the expression of glial fibrillary acidic protein (GFAP) in Muller cells were assessed by immunohistochemistry. RESULTS: Myo/Nog cells were present in the undamaged retina in low numbers. Light induced damage increased their numbers, particularly in the choroid, ganglion cell layer and outer plexiform layer. Intravitreal injection of G8-positive (G8+) cells harvested from brain mitigated all the effects of light damage examined, i.e. loss of retinal function (ERG), death of photoreceptors and the stress-induced expression of GFAP in Muller cells. Some of the transplanted G8+ cells were integrated into the retina from the vitreous. CONCLUSIONS: Myo/Nog cells are a subpopulation of cells that are present in the adult retina. They increase in number in response to light induced stress. Intravitreal injection of Myo/Nog cells was protective to the retina, in part, by reducing retinal stress as measured by the Muller cell response. These results suggest that Myo/Nog cells, or the factors they produce, are neuroprotective and may be therapeutic in neurodegenerative retinal diseases.


Subject(s)
Neuroprotection/physiology , Retina/cytology , Retina/injuries , Animals , Carrier Proteins/metabolism , Electroretinography , Glial Fibrillary Acidic Protein/metabolism , Light/adverse effects , MyoD Protein/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Rats, Sprague-Dawley , Retina/physiology
20.
Exp Eye Res ; 146: 22-25, 2016 05.
Article in English | MEDLINE | ID: mdl-26688580

ABSTRACT

Myo/Nog cells are essential for eye development in the chick embryo and respond to injury in adult tissues. These cells express mRNA for the skeletal muscle specific transcription factor MyoD, the bone morphogenetic protein (BMP) inhibitor Noggin and the cell surface protein recognized by the G8 monoclonal antibody (mAb). In this study, we determined that Myo/Nog cells are present in low numbers in the retina of the mouse eye. G8-positive Myo/Nog cells were distinguished from neuronal, Müller and microglial cells that were identified with antibodies to calretinin, Chx10, glial fibrillary acidic protein and ionized calcium binding adaptor molecule 1, respectively. In the neonatal retina, the number of Myo/Nog cells increased in parallel with cell death induced by transient exposure to hyperoxia. In this model of retinopathy of prematurity, depletion of Myo/Nog cells by intravitreal injection of the G8 mAb and complement increased cell death. These findings demonstrate that Myo/Nog cells are a distinct population of cells, not previously described in the retina, which increases in response to retinal damage and mitigate hypoxia-induced cell death.


Subject(s)
Carrier Proteins/metabolism , MyoD Protein/metabolism , Oxidative Stress , Photoreceptor Cells, Vertebrate/metabolism , Retina/metabolism , Retinopathy of Prematurity/metabolism , Animals , Cell Death , Humans , Photoreceptor Cells, Vertebrate/pathology , Retina/pathology , Retinopathy of Prematurity/diagnosis
SELECTION OF CITATIONS
SEARCH DETAIL
...