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










Publication year range
1.
Invest Ophthalmol Vis Sci ; 65(4): 43, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38683564

ABSTRACT

Purpose: Complement dysregulation is a key component in the pathogenesis of age-related macular degeneration (AMD) and related diseases such as early-onset macular drusen (EOMD). Although genetic variants of complement factor H (CFH) are associated with AMD risk, the impact of CFH and factor H-like protein 1 (FHL-1) expression on local complement activity in human retinal pigment epithelium (RPE) remains unclear. Methods: We identified a novel CFH variant in a family with EOMD and generated patient induced pluripotent stem cell (iPSC)-derived RPE cells. We assessed CFH and FHL-1 co-factor activity through C3b breakdown assays and measured complement activation by immunostaining for membrane attack complex (MAC) formation. Expression of CFH, FHL-1, local alternative pathway (AP) components, and regulators of complement activation (RCA) in EOMD RPE cells was determined by quantitative PCR, western blot, and immunostaining. Isogenic EOMD (cEOMD) RPE was generated using CRISPR/Cas9 gene editing. Results: The CFH variant (c.351-2A>G) resulted in loss of CFH and FHL-1 expression and significantly reduced CFH and FHL-1 protein expression (∼50%) in EOMD iPSC RPE cells. These cells exhibited increased MAC deposition upon exposure to normal human serum. Under inflammatory or oxidative stress conditions, CFH and FHL-1 expression in EOMD RPE cells paralleled that of controls, whereas RCA expression, including MAC formation inhibitors, was elevated. CRISPR/Cas9 correction restored CFH/FHL-1 expression and mitigated alternative pathway complement activity in cEOMD RPE cells. Conclusions: Identification of a novel CFH variant in patients with EOMD resulting in reduced CFH and FHL-1 and increased local complement activity in EOMD iPSC RPE supports the involvement of CFH haploinsufficiency in EOMD pathogenesis.


Subject(s)
Complement Factor H , Haploinsufficiency , Intracellular Signaling Peptides and Proteins , LIM Domain Proteins , Macular Degeneration , Muscle Proteins , Retinal Pigment Epithelium , Humans , Complement Factor H/genetics , Complement Factor H/metabolism , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Macular Degeneration/genetics , Macular Degeneration/metabolism , Male , Female , Induced Pluripotent Stem Cells/metabolism , Complement C3b Inactivator Proteins/genetics , Complement C3b Inactivator Proteins/metabolism , Complement Activation/genetics , Pedigree , Blotting, Western , Complement System Proteins/metabolism , Complement System Proteins/genetics , Retinal Drusen/genetics , Retinal Drusen/metabolism , Middle Aged
2.
J Biol Chem ; 299(11): 105275, 2023 11.
Article in English | MEDLINE | ID: mdl-37741457

ABSTRACT

It is known that metabolic defects in the retinal pigment epithelium (RPE) can cause degeneration of its neighboring photoreceptors in the retina, leading to retinal degenerative diseases such as age-related macular degeneration. However, how RPE metabolism supports the health of the neural retina remains unclear. The retina requires exogenous nitrogen sources for protein synthesis, neurotransmission, and energy metabolism. Using 15N tracing coupled with mass spectrometry, we found human RPE can utilize the nitrogen in proline to produce and export 13 amino acids, including glutamate, aspartate, glutamine, alanine, and serine. Similarly, we found this proline nitrogen utilization in the mouse RPE/choroid but not in the neural retina of explant cultures. Coculture of human RPE with the retina showed that the retina can take up the amino acids, especially glutamate, aspartate, and glutamine, generated from proline nitrogen in the RPE. Intravenous delivery of 15N proline in vivo demonstrated 15N-derived amino acids appear earlier in the RPE before the retina. We also found proline dehydrogenase, the key enzyme in proline catabolism is highly enriched in the RPE but not the retina. The deletion of proline dehydrogenase blocks proline nitrogen utilization in RPE and the import of proline nitrogen-derived amino acids in the retina. Our findings highlight the importance of RPE metabolism in supporting nitrogen sources for the retina, providing insight into understanding the mechanisms of the retinal metabolic ecosystem and RPE-initiated retinal degenerative diseases.


Subject(s)
Amino Acids , Retinal Pigment Epithelium , Animals , Humans , Mice , Amino Acids/metabolism , Aspartic Acid/metabolism , Glutamates/metabolism , Glutamine/metabolism , Nitrogen/metabolism , Proline/metabolism , Proline Oxidase/metabolism , Retina/metabolism , Retinal Pigment Epithelium/metabolism
3.
bioRxiv ; 2023 Jul 13.
Article in English | MEDLINE | ID: mdl-37502987

ABSTRACT

Purpose: Lipid nanoparticles (LNPs) show promise in their ability to introduce mRNA to drive protein expression in specific cell types of the mammalian eye. Here, we examined the ability of mRNA encapsulated in lipid nanoparticles (LNPs) with two distinct formulations to drive gene expression in mouse and human retina and other ocular tissues. Methods: We introduced mRNA carrying LNPs into two biological systems. Intravitreal injections were tested to deliver LNPs into the mouse eye. Human retinal pigment epithelium (RPE) and retinal explants were used to assess mRNA expression in human tissue. We analyzed specificity of expression using histology, immunofluorescence, and imaging. Results: In mice, mRNAs encoding GFP and ciliary neurotrophic factor (CNTF) were specifically expressed by Müller glia and retinal pigment epithelium (RPE). Acute inflammatory changes measured by microglia distribution (Iba-1) or interleukin-6 (IL-6) expression were not observed 6 hours post-injection. Human RPE also expressed high levels of GFP. Human retinal explants expressed GFP in cells with apical and basal processes consistent with Müller glia and in perivascular cells consistent with macrophages. Conclusions: We demonstrated the ability to reliably transfect subpopulations of retinal cells in mice eye tissues in vivo and in human ocular tissues. Of significance, intravitreal injections were sufficient to transfect the RPE in mice. To our knowledge we demonstrate delivery of mRNA using LNPs in human ocular tissues for the first time.

4.
bioRxiv ; 2023 Apr 28.
Article in English | MEDLINE | ID: mdl-37131780

ABSTRACT

It is known that metabolic defects in the retinal pigment epithelium (RPE) can cause degeneration of its neighboring photoreceptors in the retina, leading to retinal degenerative diseases such as age-related macular degeneration. However, how RPE metabolism supports the health of the neural retina remains unclear. The retina requires exogenous nitrogen sources for protein synthesis, neurotransmission, and energy metabolism. Using 15N tracing coupled with mass spectrometry, we found human RPE can utilize the nitrogen in proline to produce and export 13 amino acids, including glutamate, aspartate, glutamine, alanine and serine. Similarly, we found this proline nitrogen utilization in the mouse RPE/choroid but not in the neural retina of explant cultures. Co-culture of human RPE with the retina showed that the retina can take up the amino acids, especially glutamate, aspartate and glutamine, generated from proline nitrogen in the RPE. Intravenous delivery of 15N proline in vivo demonstrated 15N-derived amino acids appear earlier in the RPE before the retina. We also found proline dehydrogenase (PRODH), the key enzyme in proline catabolism is highly enriched in the RPE but not the retina. The deletion of PRODH blocks proline nitrogen utilization in RPE and the import of proline nitrogen-derived amino acids in the retina. Our findings highlight the importance of RPE metabolism in supporting nitrogen sources for the retina, providing insight into understanding the mechanisms of the retinal metabolic ecosystem and RPE-initiated retinal degenerative diseases.

5.
Exp Eye Res ; 215: 108899, 2022 02.
Article in English | MEDLINE | ID: mdl-34929159

ABSTRACT

Sorsby Fundus Dystrophy (SFD) is a rare form of macular degeneration that is clinically similar to age-related macular degeneration (AMD), and a histologic hallmark of SFD is a thick layer of extracellular deposits beneath the retinal pigment epithelium (RPE). Previous studies of SFD patient-induced pluripotent stem cell (iPSC) derived RPE differ as to whether these cultures recapitulate this key clinical feature by forming increased drusenoid deposits. The primary purpose of this study is to examine whether SFD patient-derived iPSC-RPE form basal deposits similar to what is found in affected family member SFD globes and to determine whether SFD iPSC RPE may be more oxidatively stressed. We performed a careful comparison of iPSC RPE from three control individuals, multiple iPSC clones from two SFD patients' iPSC RPE, and post-mortem eyes of affected SFD family members. We also examined the effect of CRISPR-Cas9 gene correction of the S204C TIMP3 mutation on RPE phenotype. Finally, targeted metabolomics with liquid chromatography and mass spectrometry analysis and stable isotope-labeled metabolite analysis were performed to determine whether SFD RPE are more oxidatively stressed. We found that SFD iPSC-RPE formed significantly more sub-RPE deposits (∼6-90 µm in height) compared to control RPE at 8 weeks. These deposits were similar in composition to the thick layer of sub-RPE deposits found in SFD family member globes by immunofluorescence staining and TEM imaging. S204C TIMP3 correction by CRISPR-Cas9 gene editing in SFD iPSC RPE cells resulted in significantly reduced basal laminar and sub-RPE calcium deposits. We detected a ∼18-fold increase in TIMP3 accumulation in the extracellular matrix (ECM) of SFD RPE, and targeted metabolomics showed that intracellular 4-hydroxyproline, a major breakdown product of collagen, is significantly elevated in SFD RPE, suggesting increased ECM turnover. Finally, SFD RPE cells have decreased intracellular reduced glutathione and were found to be more vulnerable to oxidative stress. Our findings suggest that elements of SFD pathology can be demonstrated in culture which may lead to insights into disease mechanisms.


Subject(s)
Induced Pluripotent Stem Cells , Macular Degeneration , Extracellular Matrix/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Macular Degeneration/metabolism , Retinal Pigment Epithelium/metabolism
6.
J Proteome Res ; 20(1): 909-922, 2021 01 01.
Article in English | MEDLINE | ID: mdl-32975122

ABSTRACT

Mitochondrial respiration in mammalian cells not only generates ATP to meet their own energy needs but also couples with biosynthetic pathways to produce metabolites that can be exported to support neighboring cells. However, how defects in mitochondrial respiration influence these biosynthetic and exporting pathways remains poorly understood. Mitochondrial dysfunction in retinal pigment epithelium (RPE) cells is an emerging contributor to the death of their neighboring photoreceptors in degenerative retinal diseases including age-related macular degeneration. In this study, we used targeted-metabolomics and 13C tracing to investigate how inhibition of mitochondrial respiration influences the intracellular and extracellular metabolome. We found inhibition of mitochondrial respiration strikingly influenced both the intracellular and extracellular metabolome in primary RPE cells. Intriguingly, the extracellular metabolic changes sensitively reflected the intracellular changes. These changes included substantially enhanced glucose consumption and lactate production; reduced release of pyruvate, citrate, and ketone bodies; and massive accumulation of multiple amino acids and nucleosides. In conclusion, these findings reveal a metabolic signature of nutrient consumption and release in mitochondrial dysfunction in RPE cells. Testing medium metabolites provides a sensitive and noninvasive method to assess mitochondrial function in nutrient utilization and transport.


Subject(s)
Mitochondria , Retinal Pigment Epithelium , Animals , Humans , Nutrients , Respiration , Retina/metabolism
7.
J Biol Chem ; 294(26): 10278-10289, 2019 06 28.
Article in English | MEDLINE | ID: mdl-31110046

ABSTRACT

The retinal pigment epithelium (RPE) is a monolayer of pigmented cells between the choroid and the retina. RPE dysfunction underlies many retinal degenerative diseases, including age-related macular degeneration, the leading cause of age-related blindness. To perform its various functions in nutrient transport, phagocytosis of the outer segment, and cytokine secretion, the RPE relies on an active energy metabolism. We previously reported that human RPE cells prefer proline as a nutrient and transport proline-derived metabolites to the apical, or retinal, side. In this study, we investigated how RPE utilizes proline in vivo and why proline is a preferred substrate. By using [13C]proline labeling both ex vivo and in vivo, we found that the retina rarely uses proline directly, whereas the RPE utilizes it at a high rate, exporting proline-derived mitochondrial intermediates for use by the retina. We observed that in primary human RPE cell culture, proline is the only amino acid whose uptake increases with cellular maturity. In human RPE, proline was sufficient to stimulate de novo serine synthesis, increase reductive carboxylation, and protect against oxidative damage. Blocking proline catabolism in RPE impaired glucose metabolism and GSH production. Notably, in an acute model of RPE-induced retinal degeneration, dietary proline improved visual function. In conclusion, proline is an important nutrient that supports RPE metabolism and the metabolic demand of the retina.


Subject(s)
Energy Metabolism/drug effects , Proline/administration & dosage , Retina/metabolism , Retinal Degeneration/metabolism , Retinal Pigment Epithelium/metabolism , Animals , Carbon Radioisotopes/analysis , Cell Differentiation , Humans , Male , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Oxidation-Reduction , Proline/pharmacology , Retina/drug effects , Retinal Degeneration/drug therapy , Retinal Degeneration/etiology , Retinal Pigment Epithelium/drug effects
8.
Elife ; 62017 09 13.
Article in English | MEDLINE | ID: mdl-28901286

ABSTRACT

Here we report multiple lines of evidence for a comprehensive model of energy metabolism in the vertebrate eye. Metabolic flux, locations of key enzymes, and our finding that glucose enters mouse and zebrafish retinas mostly through photoreceptors support a conceptually new model for retinal metabolism. In this model, glucose from the choroidal blood passes through the retinal pigment epithelium to the retina where photoreceptors convert it to lactate. Photoreceptors then export the lactate as fuel for the retinal pigment epithelium and for neighboring Müller glial cells. We used human retinal epithelial cells to show that lactate can suppress consumption of glucose by the retinal pigment epithelium. Suppression of glucose consumption in the retinal pigment epithelium can increase the amount of glucose that reaches the retina. This framework for understanding metabolic relationships in the vertebrate retina provides new insights into the underlying causes of retinal disease and age-related vision loss.


Subject(s)
Adaptation, Ocular , Energy Metabolism , Ependymoglial Cells/physiology , Photoreceptor Cells/physiology , Retinal Pigment Epithelium/physiology , Animals , Ependymoglial Cells/metabolism , Glucose/metabolism , Humans , Lactates/metabolism , Mice , Photoreceptor Cells/metabolism , Retinal Pigment Epithelium/metabolism , Zebrafish
9.
J Biol Chem ; 292(31): 12895-12905, 2017 08 04.
Article in English | MEDLINE | ID: mdl-28615447

ABSTRACT

Metabolite transport is a major function of the retinal pigment epithelium (RPE) to support the neural retina. RPE dysfunction plays a significant role in retinal degenerative diseases. We have used mass spectrometry with 13C tracers to systematically study nutrient consumption and metabolite transport in cultured human fetal RPE. LC/MS-MS detected 120 metabolites in the medium from either the apical or basal side. Surprisingly, more proline is consumed than any other nutrient, including glucose, taurine, lipids, vitamins, or other amino acids. Besides being oxidized through the Krebs cycle, proline is used to make citrate via reductive carboxylation. Citrate, made either from 13C proline or from 13C glucose, is preferentially exported to the apical side and is taken up by the retina. In conclusion, RPE cells consume multiple nutrients, including glucose and taurine, but prefer proline, and they actively synthesize and export metabolic intermediates to the apical side to nourish the outer retina.


Subject(s)
Proline/metabolism , Retina/metabolism , Retinal Pigment Epithelium/metabolism , Animals , Biological Transport , Carbon Isotopes , Cell Polarity , Cells, Cultured , Citric Acid/metabolism , Citric Acid Cycle , Coculture Techniques , Embryo, Mammalian/cytology , Glucose/metabolism , Humans , Kinetics , Metabolomics/methods , Mice , Retina/cytology , Retina/enzymology , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/enzymology , Taurine/metabolism , Tissue Culture Techniques
10.
Transl Vis Sci Technol ; 6(3): 4, 2017 May.
Article in English | MEDLINE | ID: mdl-28516002

ABSTRACT

PURPOSE: Previous studies have demonstrated the ability of retinal cells derived from human embryonic stem cells (hESCs) to survive, integrate into the host retina, and mediate light responses in murine mouse models. Our aim is to determine whether these cells can also survive and integrate into the retina of a nonhuman primate, Saimiri sciureus, following transplantation into the subretinal space. METHODS: hESCs were differentiated toward retinal neuronal fates using our previously published technique and cultured for 60 to 70 days. Differentiated cells were further treated with 20 µM N-[N-(3,5-Difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester (DAPT) for a period of 5 days immediately prior to subretinal transplantation. Differentiated cells were labeled with a lentivirus expressing GFP. One million cells (10,000 cells/µL) were injected into the submacular space into a squirrel monkey eye, using an ab externo technique. RESULTS: RetCam imaging demonstrated the presence and survival of human donor cells 3 months after transplantation in the S. sciureus eye. Injected cells consolidated in the temporal macula. GFP+ axonal projections were observed to emanate from the central consolidation of cells at 1 month, with some projecting into the optic nerve by 3 months after transplantation. CONCLUSIONS: Human ES cell-derived retinal neurons injected into the submacular space of a squirrel monkey survive at least 3 months postinjection without immunosuppression. Some donor cells appeared to integrate into the host inner retina, and numerous donor axonal projections were noted throughout, with some projecting into the optic nerve. TRANSLATIONAL RELEVANCE: These data illustrate the feasibility of hESC-derived retinal cell replacement in the nonhuman primate eye.

11.
Proc Natl Acad Sci U S A ; 113(51): 14710-14715, 2016 12 20.
Article in English | MEDLINE | ID: mdl-27911769

ABSTRACT

The retinal pigment epithelium (RPE) is a monolayer of pigmented cells that requires an active metabolism to maintain outer retinal homeostasis and compensate for oxidative stress. Using 13C metabolic flux analysis in human RPE cells, we found that RPE has an exceptionally high capacity for reductive carboxylation, a metabolic pathway that has recently garnered significant interest because of its role in cancer cell survival. The capacity for reductive carboxylation in RPE exceeds that of all other cells tested, including retina, neural tissue, glial cells, and a cancer cell line. Loss of reductive carboxylation disrupts redox balance and increases RPE sensitivity to oxidative damage, suggesting that deficiencies of reductive carboxylation may contribute to RPE cell death. Supporting reductive carboxylation by supplementation with an NAD+ precursor or its substrate α-ketoglutarate or treatment with a poly(ADP ribose) polymerase inhibitor protects reductive carboxylation and RPE viability from excessive oxidative stress. The ability of these treatments to rescue RPE could be the basis for an effective strategy to treat blinding diseases caused by RPE dysfunction.


Subject(s)
Carbon/chemistry , Eye/embryology , Ketoglutaric Acids/chemistry , Macular Degeneration/metabolism , Retinal Pigment Epithelium/embryology , Retinal Pigment Epithelium/metabolism , Aged, 80 and over , Animals , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Cell Survival , Fatty Acids/chemistry , Female , HeLa Cells , Humans , Induced Pluripotent Stem Cells/metabolism , Isocitrate Dehydrogenase/metabolism , Macular Degeneration/pathology , Mice , NAD/chemistry , Neoplasms/metabolism , Neoplasms/pathology , Oxidation-Reduction , Oxidative Stress , Oxygen/chemistry , Poly(ADP-ribose) Polymerases/metabolism
12.
Immunobiology ; 218(12): 1468-76, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23735481

ABSTRACT

Protein-bound polysaccharide-K (PSK) is a hot water extract from Trametes versicolor mushroom. It has been used traditionally in Asian countries for its immune stimulating and anti-cancer effects. We have recently found that PSK can activate Toll-like receptor 2 (TLR2). TLR2 is highly expressed on dendritic cells (DC), so the current study was undertaken to evaluate the effect of PSK on DC activation and the potential of using PSK as a vaccine adjuvant. In vitro experiments using mouse bone marrow-derived DC (BMDC) demonstrated that PSK induces DC maturation as shown by dose-dependent increase in the expression of CD80, CD86, MHCII, and CD40. PSK also induces the production of multiple inflammatory cytokines by DC, including IL-12, TNF-α, and IL-6, at both mRNA and protein levels. In vivo experiments using PSK as an adjuvant to OVAp323-339 vaccine showed that PSK as adjuvant leads to enlarged draining lymph nodes with higher number of activated DC. PSK also stimulates proliferation of OVA-specific T cells, and induces T cells that produce multiple cytokines, IFN-γ, IL-2, and TNF-α. Altogether, these results demonstrate the ability of PSK to activate DC in vitro and in vivo and the potential of using PSK as a novel vaccine adjuvant.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Dendritic Cells/drug effects , Proteoglycans/administration & dosage , T-Lymphocytes/immunology , Vaccines, Subunit , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Dendritic Cells/immunology , Inflammation Mediators/metabolism , Lymphocyte Activation/drug effects , Mice , Mice, Inbred BALB C , Mice, Transgenic , Ovalbumin/immunology , Peptide Fragments/immunology , T-Cell Antigen Receptor Specificity , Trametes/immunology , Up-Regulation/drug effects
13.
Expert Rev Clin Pharmacol ; 4(2): 275-89, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21643519

ABSTRACT

Toll-like receptor (TLR) ligation activates both the innate and adaptive immune systems, and plays an important role in antiviral and anti-tumor immunity. Therefore, a significant amount of effort has been devoted to exploit the therapeutic potential of TLR agonists. Depending on the therapeutic purpose, either as adjuvants to vaccine, chemotherapy or standalone therapy, TLR agonists have been administered via different routes. Both preclinical and clinical studies have suggested that the route of administration has significant effects on pharmacokinetics, and that understanding these effects is critical to the success of TLR agonist drug development. This article will summarize the pharmacokinetics of TLR agonists with different administration routes, with an emphasis on clinical studies of TLR ligands in oncologic applications.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/pharmacokinetics , Immune System/drug effects , Toll-Like Receptors/agonists , Adjuvants, Immunologic/metabolism , Animals , Clinical Trials as Topic/methods , Drug Delivery Systems/methods , Humans , Immune System/immunology , Immune System/metabolism , Ligands , Signal Transduction/drug effects , Signal Transduction/immunology , Toll-Like Receptors/metabolism
14.
Neurosci Lett ; 388(2): 106-11, 2005 Nov 11.
Article in English | MEDLINE | ID: mdl-16039063

ABSTRACT

The transmissible spongiform encephalopathies (TSEs) are fatal neurodegenerative diseases. A primary therapeutic target for TSE intervention has been a protease-resistant form of prion protein known as PrP(Sc) or PrP-res. In vitro testing of mouse scrapie-infected cell cultures has identified many PrP-res inhibitors that also have activity in vivo. Here we identify 32 new inhibitors of two strains of mouse scrapie PrP-res. Furthermore, to investigate the species-specificity of these and other PrP-res inhibitors, we have developed a high-throughput cell culture assay based on Rov9 cells chronically-infected with sheep scrapie. Of 32 inhibitors of murine PrP-res that were also tested in the Rov9 cells, only six showed inhibitory activity against sheep PrP-res. The three most potent inhibitors of both murine and ovine PrP-res formation (with 50% inhibition at < or =5 microM) were tannic acid, pentosan polysulfate and Fe(III) deuteroporphyrin 2,4-bisethyleneglycol. The latter two have anti-mouse scrapie activity in vivo. These results identify new inhibitors of murine and ovine PrP-res formation and reinforce the idea that compounds effective against PrP-res from one species or strain cannot be assumed to be active against others.


Subject(s)
PrPSc Proteins/antagonists & inhibitors , Scrapie/drug therapy , Scrapie/metabolism , Tannins/pharmacology , Animals , Cell Line , Deuteroporphyrins/pharmacology , Enzyme Inhibitors/pharmacology , Epithelial Cells/cytology , Ferric Compounds/pharmacology , Mice , Pentosan Sulfuric Polyester/pharmacology , PrPSc Proteins/metabolism , Rabbits , Sheep , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...