Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
J Biomed Opt ; 28(11): 116002, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-38078154

RESUMO

Significance: Over 100 monoclonal antibodies have been approved by the U.S. Food and Drug Administration (FDA) for clinical use; however, a paucity of knowledge exists regarding the injection site behavior of these formulated therapeutics, particularly the effect of antibody, formulation, and tissue at the injection site. A deeper understanding of antibody behavior at the injection site, especially on blood oxygenation through imaging, will help design improved versions of the therapeutics for a wide range of diseases. Aim: The aim of this research is to understand the dynamics of monoclonal antibodies at the injection site as well as how the antibody itself affects the functional characteristics of the injection site [e.g., blood oxygen saturation (sO2)]. Approach: We employed triple-wavelength equipped functional photoacoustic imaging to study the dynamics of dye-labeled and unlabeled monoclonal antibodies at the site of injection in a mouse ear. We injected a near-infrared dye-labeled (and unlabeled) human IgG4 isotype control antibody into the subcutaneous space in mouse ears to analyze the injection site dynamics and quantify molecular movement, as well as its effect on local hemodynamics. Results: We performed pharmacokinetic studies of the antibody in different regions of the mouse body to show that dye labeling does not alter the pharmacokinetic characteristics of the antibody and that mouse ear is a viable model for these initial studies. We explored the movement of the antibody in the interstitial space to show that the bolus area grows by ∼300% over 24 h. We discovered that injection of the antibody transiently reduces the local sO2 levels in mice after prolonged anesthesia without affecting the total hemoglobin content and oxygen extraction fraction. Conclusions: This finding on local oxygen saturation opens a new avenue of study on the functional effects of monoclonal antibody injections. We also show the suitability of the mouse ear model to study antibody dynamics through high-resolution imaging techniques. We quantified the movement of antibodies at the injection site caused by the interstitial fluid, which could be helpful for designing antibodies with tailored absorption speeds in the future.


Assuntos
Anestesia , Técnicas Fotoacústicas , Camundongos , Humanos , Animais , Anticorpos Monoclonais , Tela Subcutânea , Imunoglobulina G
2.
Mol Metab ; 62: 101522, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35671972

RESUMO

OBJECTIVE: Ultra-rapid insulin formulations control postprandial hyperglycemia; however, inadequate understanding of injection site absorption mechanisms is limiting further advancement. We used photoacoustic imaging to investigate the injection site dynamics of dye-labeled insulin lispro in the Humalog® and Lyumjev® formulations using the murine ear cutaneous model and correlated it with results from unlabeled insulin lispro in pig subcutaneous injection model. METHODS: We employed dual-wavelength optical-resolution photoacoustic microscopy to study the absorption and diffusion of the near-infrared dye-labeled insulin lispro in the Humalog and Lyumjev formulations in mouse ears. We mathematically modeled the experimental data to calculate the absorption rate constants and diffusion coefficients. We studied the pharmacokinetics of the unlabeled insulin lispro in both the Humalog and Lyumjev formulations as well as a formulation lacking both the zinc and phenolic preservative in pigs. The association state of insulin lispro in each of the formulations was characterized using SV-AUC and NMR spectroscopy. RESULTS: Through experiments using murine and swine models, we show that the hexamer dissociation rate of insulin lispro is not the absorption rate-limiting step. We demonstrated that the excipients in the Lyumjev formulation produce local tissue expansion and speed both insulin diffusion and microvascular absorption. We also show that the diffusion of insulin lispro at the injection site drives its initial absorption; however, the rate at which the insulin lispro crosses the blood vessels is its overall absorption rate-limiting step. CONCLUSIONS: This study provides insights into injection site dynamics of insulin lispro and the impact of formulation excipients. It also demonstrates photoacoustic microscopy as a promising tool for studying protein therapeutics. The results from this study address critical questions around the subcutaneous behavior of insulin lispro and the formulation excipients, which could be useful to make faster and better controlled insulin formulations in the future.


Assuntos
Insulina de Ação Curta , Técnicas Fotoacústicas , Animais , Excipientes , Hipoglicemiantes/química , Insulina , Insulina Lispro , Camundongos , Suínos
3.
Bioorg Med Chem ; 32: 115942, 2021 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-33461147

RESUMO

The neonatal Fc receptor (FcRn) represents a transport system with the potential to facilitate absorption of biologics across the gastrointestinal barrier. How biologics interact with FcRn to enable their gastrointestinal absorption, and how these interactions might be optimized in a biological therapeutic are not well understood. Thus, we studied the absorption of Fc molecules from the intestine using three IgG4-derived Fc variants with different, pH-dependent FcRn binding and release profiles. Using several different intestinal models, we consistently observed that FcRn binding affinity correlated with transcytosis. Our findings support targeting FcRn to enable intestinal absorption of biologics and highlight additional strategic considerations for future work.


Assuntos
Antígenos de Histocompatibilidade Classe I/química , Fragmentos Fc das Imunoglobulinas/química , Receptores Fc/química , Sítios de Ligação , Células Cultivadas , Absorção Gastrointestinal , Células HEK293 , Antígenos de Histocompatibilidade Classe I/genética , Humanos , Concentração de Íons de Hidrogênio , Absorção Intestinal , Receptores Fc/genética
4.
Transplantation ; 101(3): 517-523, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27379553

RESUMO

BACKGROUND: The rapidly improving tools of genetic engineering may make it possible to overcome the humoral immune barrier that prevents xenotransplantation. We hypothesize that levels of human antibody binding to donor tissues from swine must approximate the antibody binding occurring in allotransplantation. It is uncertain if this is an attainable goal. Here we perform an initial analysis of this issue by comparing human antibody binding to red blood cells (RBC) isolated from knockout swine and to allogeneic or autologous human RBC. METHODS: Human sera were incubated with RBC isolated from various genetically engineered swine or from humans. The level of IgG and IgM binding to these cells were compared using either flow cytometry or a novel mass spectrometric assay. RESULTS: Mass spectroscopic quantitation of human antibody binding demonstrated that as few as 3 gene inactivations can reduce the levels human antibody binding to swine RBC that is as low as autologous human RBC. Flow cytometry showed that RBC from 2-gene knockout swine exhibited less human antibody binding than human blood group O allogeneic RBC in 22% of tested sera. Deletion of a third gene from pigs resulted in 30% of human samples having less IgG and IgM RBC xenoreactivity than alloreactivity. CONCLUSIONS: Xenoantigenicity of swine RBC can be eliminated via gene disruption. These results suggest that the gene knockout approach may be able reduce antigenicity in other pig tissues to levels that enable the xenotransplantation humoral barrier to be overcome.


Assuntos
Antígenos Heterófilos/genética , Antígenos Heterófilos/imunologia , Eritrócitos/imunologia , Técnicas de Inativação de Genes , Histocompatibilidade , Suínos/imunologia , Animais , Animais Geneticamente Modificados , Antígenos Heterófilos/sangue , Sítios de Ligação de Anticorpos , Citometria de Fluxo , Sobrevivência de Enxerto , Humanos , Imunidade Humoral , Isoanticorpos/sangue , Isoanticorpos/imunologia , Ligação Proteica , Suínos/sangue , Suínos/genética , Espectrometria de Massas em Tandem , Tolerância ao Transplante , Transplante Heterólogo
5.
Transplantation ; 100(3): 533-7, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26906938

RESUMO

BACKGROUND: Disrupting the porcine GGTA1 and CMAH genes [double knockout (DKO)] that produce the gal-α(1,3)-gal and N-glycolylneuraminic acid xenoantigens reduces human antibody binding to porcine peripheral blood mononuclear cells. It is important to examine rejection pathways at an organ-specific level. The object of this study is to evaluate the human preformed antibody reactivity against DKO renal microvascular endothelial cells (RMEC) in vitro. METHODS: Characteristics of DKO RMEC were analyzed using flow cytometry. Human IgG/M binding to primary RMEC, immortalized RMEC (iRMEC), and iRMEC-deficient in B4GALNT2 genes were examined using flow cytometric crossmatch assay. RESULTS: Porcine RMEC expressed gal-α(1,3)-gal, N-glycolylneuraminic acid, and Dolichos biflorus agglutinin glycans recognized by human preexisting antibodies in humans. Antigenicity of DKO RMEC was lower than GGTA1 KO RMEC. The disruption of B4GALNT2 gene in DKO iRMEC further reduced human IgG/IgM binding. CONCLUSIONS: Silencing the porcine GGTA1, CMAH, and B4GALNT2 genes is an effective strategy to reduce human preformed antibody binding to RMEC. Porcine RMEC will be a useful reagent for the further study of xenoimmunology.


Assuntos
Antígenos Heterófilos/imunologia , Células Endoteliais/imunologia , Rim/irrigação sanguínea , Microvasos/imunologia , Animais , Animais Geneticamente Modificados , Antígenos Heterófilos/metabolismo , Células Cultivadas , Células Endoteliais/metabolismo , Galactosiltransferases/deficiência , Galactosiltransferases/genética , Galactosiltransferases/imunologia , Técnicas de Inativação de Genes , Sobrevivência de Enxerto , Humanos , Imunoglobulina G/imunologia , Imunoglobulina G/metabolismo , Imunoglobulina M/imunologia , Imunoglobulina M/metabolismo , Microvasos/citologia , Microvasos/metabolismo , Oxigenases de Função Mista/deficiência , Oxigenases de Função Mista/genética , Oxigenases de Função Mista/imunologia , N-Acetilgalactosaminiltransferases/deficiência , N-Acetilgalactosaminiltransferases/genética , N-Acetilgalactosaminiltransferases/imunologia , Fenótipo , Suínos , Transfecção
6.
Transplantation ; 100(3): 571-6, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26906939

RESUMO

BACKGROUND: A profound thrombocytopenia limits hepatic xenotransplantation in the pig-to-primate model. Porcine livers also have shown the ability to phagocytose human platelets in the absence of immune-mediated injury. Recently, inactivation of the porcine ASGR1 gene has been shown to decrease this phenomenon. Inactivating GGTA1 and CMAH genes has reduced the antibody-mediated barrier to xenotransplantation; herein, we describe the effect that these modifications have on xenogeneic consumption of human platelets in the absence of immune-mediated graft injury. METHODS: Wild type (WT), ASGR1, GGTA1, and GGTA1CMAH knockout pigs were compared for their xenogeneic hepatic consumption of human platelets. An in vitro assay was established to measure the association of human platelets with liver sinusoidal endothelial cells (LSECs) by immunohistochemistry. Perfusion models were used to measure human platelet uptake in livers from WT, ASGR1, GGTA1, and GGTA1 CMAH pigs. RESULTS: GGTA1, CMAH LSECs exhibited reduced levels of human platelet binding in vitro when compared with GGTA1 and WT LSECs. In a continuous perfusion model, GGTA1 CMAH livers consumed fewer human platelets than GGTA1 and WT livers. GGTA1 CMAH livers also consumed fewer human platelets than ASGR1 livers in a single-pass model. CONCLUSIONS: Silencing the porcine carbohydrate genes necessary to avoid antibody-mediated rejection in a pig-to-human model also reduces the xenogeneic consumption of human platelets by the porcine liver. The combination of these genetic modifications may be an effective strategy to limit the thrombocytopenia associated with pig-to-human hepatic xenotransplantation.


Assuntos
Plaquetas/metabolismo , Galactosiltransferases/genética , Fígado/metabolismo , Oxigenases de Função Mista/genética , Fagocitose , Trombocitopenia/prevenção & controle , Animais , Animais Geneticamente Modificados , Anticorpos Heterófilos/imunologia , Anticorpos Heterófilos/metabolismo , Antígenos Heterófilos/imunologia , Antígenos Heterófilos/metabolismo , Receptor de Asialoglicoproteína/deficiência , Receptor de Asialoglicoproteína/genética , Receptor de Asialoglicoproteína/imunologia , Plaquetas/imunologia , Células Cultivadas , Galactosiltransferases/deficiência , Galactosiltransferases/imunologia , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/prevenção & controle , Sobrevivência de Enxerto , Xenoenxertos , Humanos , Fígado/imunologia , Oxigenases de Função Mista/deficiência , Oxigenases de Função Mista/imunologia , Adesividade Plaquetária , Suínos , Trombocitopenia/sangue , Trombocitopenia/imunologia , Trombocitopenia/metabolismo , Fatores de Tempo
7.
Xenotransplantation ; 22(3): 203-10, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25728617

RESUMO

BACKGROUND: The lethal thrombocytopenia that accompanies liver xenotransplantation is a barrier to clinical application. Human platelets are bound by the asialoglycoprotein receptor (ASGR) on pig sinusoidal endothelial cells and phagocytosed. Inactivation of the ASGR1 gene in donor pigs may prevent xenotransplantation-induced thrombocytopenia. METHODS: Transcription activator-like effector nucleases (TALENs) were targeted to the ASGR1 gene in pig liver-derived cells. ASGR1 deficient pig cells were used for somatic cell nuclear transfer (SCNT). ASGR1 knock out (ASGR1-/-) fetal fibroblasts were used to produce healthy ASGR1 knock out piglets. Human platelet uptake was measured in ASGR1+/+ and ASGR1-/- livers. RESULTS: Targeted disruption of the ASGR1 gene with TALENs eliminated expression of the receptor. ASGR1-/- livers phagocytosed fewer human platelets than domestic porcine livers during perfusion. CONCLUSIONS: The use of TALENs in liver-derived cells followed by SCNT enabled the production of healthy homozygous ASGR1 knock out pigs. Livers from ASGR1-/- pigs exhibit decreased human platelet uptake. Deletion of the ASGR1 gene is a viable strategy to diminish platelet destruction in pig-to-human xenotransplantation.


Assuntos
Receptor de Asialoglicoproteína/metabolismo , Plaquetas/metabolismo , Fígado/citologia , Transplante Heterólogo , Animais , Receptor de Asialoglicoproteína/genética , Células Endoteliais/metabolismo , Técnicas de Inativação de Genes/métodos , Hepatócitos/metabolismo , Humanos , Técnicas de Transferência Nuclear , Suínos , Trombocitopenia/imunologia
8.
Xenotransplantation ; 22(1): 20-31, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25178170

RESUMO

BACKGROUND: Manipulating the pig genome to increase compatibility with human biology may facilitate the clinical application of xenotransplantation. Genetic modifications to pig cells have been made by sequential recombination in fetal fibroblasts and liver-derived cells followed by cross-breeding or somatic cell nuclear transfer. The generation of pigs for research or organ donation by these methods is slow, expensive and requires technical expertise. A novel system incorporating the bacterial nuclease Cas9 and single-guide RNA targeting a 20 nucleotide site within a gene can be expressed from a single plasmid leading to a double-strand break and gene disruption. Coexpression of multiple unique single-guide RNA can modify several genetic loci in a single step. We describe a process for increasing the efficiency of selecting cells with multiple genetic modifications. METHODS: We used the CRISPR/Cas system to target the GGTA1, CMAH and putative iGb3S genes in pigs that have been naturally deleted in humans. Cells lacking galactose α-1,3 galactose (α-Gal) were negatively selected by an IB4 lectin/magnetic bead. α-Gal negative multiplexed single-guide RNA-treated cells were used for somatic cell nuclear transfer (SCNT) and transferred to fertile sows. We examined the levels of α-Gal and Neu5Gc expression of 32 day fetuses and piglets and analyzed the targeted genes by DNA sequencing. RESULTS: Liver-derived cells treated with multiple single-guide RNA and selected for an α-Gal null phenotype were significantly more likely to also carry mutations in simultaneously targeted genes. Multiplex single-guide RNA-treated cells used directly for SCNT without further genetic selection produced piglets with deletions in the targeted genes but also created double- and triple-gene KO variations. CRISPR/Cas-treated cells grew normally and yielded normal liters of healthy piglets via somatic cell nuclear transfer. CONCLUSIONS: The CRISPR/Cas system allows targeting of multiple genes in a single reaction with the potential to create pigs of one genetic strain or multiple genetic modifications in a single pregnancy. The application of this phenotypic selection strategy with multiplexed sgRNA and the Cas9 nuclease has accelerated our ability to produce and evaluate pigs important to xenotransplantation.


Assuntos
Sistemas CRISPR-Cas , Galactosiltransferases/genética , Técnicas de Inativação de Genes , Oxigenases de Função Mista/genética , Técnicas de Transferência Nuclear , RNA Guia de Cinetoplastídeos/genética , Sus scrofa/genética , Animais , Antígenos Heterófilos/genética , Biotinilação , Feminino , Deleção de Genes , Vetores Genéticos , Hepatócitos/citologia , Separação Imunomagnética , Fenótipo , Lectinas de Plantas/metabolismo , Gravidez , Estreptavidina , Suínos
9.
J Immunol ; 193(11): 5751-7, 2014 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-25339675

RESUMO

Pigs are emerging as important large animal models for biomedical research, and they may represent a source of organs for xenotransplantation. The MHC is pivotal to the function of the immune system in health and disease, and it is particularly important in infection and transplant rejection. Pigs deficient in class I MHC could serve as important reagents to study viral immunity as well as allograft and xenograft rejection. In this study, we report the creation and characterization of class I MHC knockout pigs using the Cas9 nuclease and guide RNAs. Pig fetal fibroblasts were genetically engineered using Cas9 and guide RNAs, and class I MHC(-) cells were then used as nuclear donors for somatic cell nuclear transfer. We produced three piglets devoid of all cell surface class I proteins. Although these animals have reduced levels of CD4(-)CD8(+) T cells in peripheral blood, the pigs appear healthy and are developing normally. These pigs are a promising reagent for immunological research.


Assuntos
Fibroblastos/fisiologia , Técnicas de Silenciamento de Genes/métodos , Antígenos de Histocompatibilidade Classe I/genética , Suínos/imunologia , Linfócitos T/imunologia , Transplante Heterólogo , Viroses/imunologia , Animais , Células Cultivadas , Endonucleases/metabolismo , Engenharia Genética , Rejeição de Enxerto , Modelos Animais , Técnicas de Transferência Nuclear , RNA Guia de Cinetoplastídeos/genética , Suínos/genética
10.
Xenotransplantation ; 20(1): 27-35, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23384142

RESUMO

BACKGROUND: Clinical xenotransplantation is not possible because humans possess antibodies that recognize antigens on the surface of pig cells. Galα-1,3-Gal (Gal) and N-glycolylneuraminic acid (Neu5Gc) are two known xenoantigens. METHODS: We report the homozygous disruption of the α1, 3-galactosyltransferase (GGTA1) and the cytidine monophosphate-N-acetylneuraminic acid hydroxylase (CMAH) genes in liver-derived female pig cells using zinc-finger nucleases (ZFNs). Somatic cell nuclear transfer (SCNT) was used to produce healthy cloned piglets from the genetically modified liver cells. Antibody-binding and antibody-mediated complement-dependent cytotoxicity assays were used to examine the immunoreactivity of pig cells deficient in Neu5Gc and Gal. RESULTS: This approach enabled rapid production of a pig strain deficient in multiple genes without extensive breeding protocols. Immune recognition studies showed that pigs lacking both CMAH and GGTA1 gene activities reduce the humoral barrier to xenotransplantation, further than pigs lacking only GGTA1. CONCLUSIONS: This technology will accelerate the development of pigs for xenotransplantation research.


Assuntos
Dissacarídeos/imunologia , Ácidos Neuramínicos/imunologia , Sus scrofa/genética , Sus scrofa/imunologia , Transplante Heterólogo/imunologia , Animais , Anticorpos Heterófilos/metabolismo , Citotoxicidade Celular Dependente de Anticorpos , Antígenos Heterófilos/imunologia , Antígenos Heterófilos/metabolismo , Sequência de Bases , Células Cultivadas , DNA/genética , Dissacarídeos/deficiência , Feminino , Galactosiltransferases/deficiência , Galactosiltransferases/genética , Técnicas de Inativação de Genes/métodos , Humanos , Leucócitos Mononucleares/imunologia , Oxigenases de Função Mista/deficiência , Oxigenases de Função Mista/genética , Ácidos Neuramínicos/metabolismo , Sus scrofa/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA