Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
1.
Biomacromolecules ; 22(7): 2850-2863, 2021 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-34156837

RESUMEN

Over the past decade, extensive optimization of polymeric cell-penetrating peptide (CPP) mimics (CPPMs) by our group has generated a substantial library of broadly effective carriers which circumvent the need for covalent conjugation often required by CPPs. In this study, design rules learned from CPPM development were applied to reverse-engineer the first library of simple amphiphilic block copolypeptides for non-covalent protein delivery, namely, poly(alanine-block-arginine), poly(phenylalanine-block-arginine), and poly(tryptophan-block-arginine). This new CPP library was screened for enhanced green fluorescent protein and Cre recombinase delivery alongside a library of CPPMs featuring equivalent side-chain configurations. Due to the added hydrophobicity imparted by the polymer backbone as compared to the polypeptide backbone, side-chain functionality was not a universal predictor of carrier performance. Rather, overall carrier hydrophobicity predicted the top performers for both internalization and activity of protein cargoes, regardless of backbone identity. Furthermore, comparison of protein uptake and function revealed carriers which facilitated high gene recombination despite remarkably low Cre internalization, leading us to formalize the concept of intracellular availability (IA) of the delivered cargo. IA, a measure of cargo activity per quantity of cargo internalized, provides valuable insight into the physical relationship between cellular internalization and bioavailability, which can be affected by bottlenecks such as endosomal escape and cargo release. Importantly, carriers with maximal IA existed within a narrow hydrophobicity window, more hydrophilic than those exhibiting maximal cargo uptake. Hydrophobicity may be used as a scaffold-independent predictor of protein uptake, function, and IA, enabling identification of new, effective carriers which would be overlooked by uptake-based screening methods.


Asunto(s)
Péptidos de Penetración Celular , Transporte Biológico , Péptidos de Penetración Celular/metabolismo , Interacciones Hidrofóbicas e Hidrofílicas , Polímeros , Transporte de Proteínas
2.
Mol Ther ; 28(10): 2220-2236, 2020 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-32592691

RESUMEN

T cell receptor signaling, together with cytokine-induced signals, can differentially regulate RNA processing to influence T helper versus regulatory T cell fate. Protein kinase C family members have been shown to function in alternative splicing and RNA processing in various cell types. T cell-specific protein kinase C theta, a molecular regulator of T cell receptor downstream signaling, has been shown to phosphorylate splicing factors and affect post-transcriptional control of T cell gene expression. In this study, we explored how using a synthetic cell-penetrating peptide mimic for intracellular anti-protein kinase C theta delivery fine-tunes differentiation of induced regulatory T cells through its differential effects on RNA processing. We identified protein kinase C theta signaling as a critical modulator of two key RNA regulatory factors, heterogeneous nuclear ribonucleoprotein L (hnRNPL) and protein-l-isoaspartate O-methyltransferase-1 (PCMT1), and loss of protein kinase C theta function initiated a "switch" in post-transcriptional organization in induced regulatory T cells. More interestingly, we discovered that protein-l-isoaspartate O- methyltransferase-1 acts as an instability factor in induced regulatory T cells, by methylating the forkhead box P3 (FOXP3) promoter. Targeting protein-l-isoaspartate O-methyltransferase-1 using a cell-penetrating antibody revealed an efficient means of modulating RNA processing to confer a stable regulatory T cell phenotype.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica , Ribonucleoproteína Heterogénea-Nuclear Grupo L/metabolismo , Proteína D-Aspartato-L-Isoaspartato Metiltransferasa/genética , Proteína Quinasa C-theta/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Péptidos de Penetración Celular/química , Péptidos de Penetración Celular/metabolismo , Péptidos de Penetración Celular/farmacología , Factores de Transcripción Forkhead/genética , Regiones Promotoras Genéticas , Unión Proteica , Proteína D-Aspartato-L-Isoaspartato Metiltransferasa/metabolismo , Estabilidad Proteica , Transducción de Señal
3.
Mol Ther ; 28(9): 1987-2006, 2020 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-32492367

RESUMEN

Regulatory T cells maintain immunological tolerance and dampen inflammatory responses. Administering regulatory T cells can prevent the immune-mediated tissue destruction of graft-versus-host disease, which frequently accompanies hematopoietic stem cell transfer. Neutralizing the T cell-specific kinase, protein kinase C theta, which promotes T cell effector functions and represses regulatory T cell differentiation, augments regulatory T cell immunosuppression and stability. We used a synthetic, cell-penetrating peptide mimic to deliver antibodies recognizing protein kinase C theta into primary human CD4 T cells. When differentiated ex vivo into induced regulatory T cells, treated cells expressed elevated levels of the regulatory T cell transcriptional regulator forkhead box P3, the surface-bound immune checkpoint receptor programmed death receptor-1, and pro-inflammatory interferon gamma, previously ascribed to a specific population of stable, highly suppressive human induced regulatory T cells. The in vitro suppressive capacity of these induced regulatory T cells was 10-fold greater than that of T cells differentiated without antibody delivery. When administered at the time of graft-versus-host disease induction, using a humanized mouse model, antibody-treated regulatory T cells were superior to non-treated T cells in attenuating lethal outcomes. This antibody delivery approach may overcome obstacles currently encountered using patient-derived regulatory T cells as a cell-based therapy for immune modulation.


Asunto(s)
Traslado Adoptivo/métodos , Anticuerpos/inmunología , Anticuerpos/farmacología , Péptidos de Penetración Celular , Enfermedad Injerto contra Huésped/terapia , Tolerancia Inmunológica/efectos de los fármacos , Líquido Intracelular/inmunología , Proteína Quinasa C-theta/inmunología , Linfocitos T Reguladores/inmunología , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Factores de Transcripción Forkhead/metabolismo , Enfermedad Injerto contra Huésped/inmunología , Humanos , Tolerancia Inmunológica/inmunología , Interferón gamma/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Receptor de Muerte Celular Programada 1/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Resultado del Tratamiento
4.
J Biol Chem ; 294(29): 11276-11285, 2019 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-31167792

RESUMEN

Presenilins 1 and 2 (PS1 and 2) are the catalytic subunits of γ-secretase, a multiprotein protease that cleaves amyloid protein precursor and other type I transmembrane proteins. Previous studies with mouse models or cells have indicated differences in PS1 and PS2 functions. We have recently reported that clinical γ-secretase inhibitors (GSIs), initially developed to manage Alzheimer's disease and now being considered for other therapeutic interventions, are both pharmacologically and functionally distinct. Here, using CRISPR/Cas9-based gene editing, we established human HEK 293T cell lines in which endogenous PS1, PS2, or both have been knocked out. Using these knockout lines to examine differences in PS1- and PS2-mediated cleavage events, we confirmed that PS2 generates more intracellular ß-amyloid than does PS1. Moreover, we observed subtle differences in PS1- and PS2-mediated cleavages of select substrates. In exploring the question of whether differences in activity among clinical GSIs could be attributed to differential inhibition of PS1 or PS2, we noted that select GSIs inhibit PS1 and PS2 activities on specific substrates with slightly different potencies. We also found that endoproteolysis of select PS1 FAD-linked variants in human cells is more efficient than what has been previously reported for mouse cell lines. Overall, these results obtained with HEK293T cells suggest that selective PS1 or PS2 inhibition by a given GSI does not explain the previously observed differences in functional and pharmacological properties among various GSIs.


Asunto(s)
Presenilina-1/fisiología , Presenilina-2/fisiología , Enfermedad de Alzheimer/genética , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Sistemas CRISPR-Cas , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Hidrólisis , Ratones , Presenilina-1/genética , Presenilina-2/genética , Especificidad por Sustrato
5.
Mol Ther ; 27(8): 1436-1451, 2019 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-31138510

RESUMEN

Acute graft-versus-host disease is a frequent complication associated with allogeneic hematopoietic stem cell transplantation. Patients that become refractory to initial steroid treatment have a poor prognosis. apceth-201 consists of human allogeneic mesenchymal stromal cells, engineered by lentiviral transduction to express the protease inhibitor alpha-1 antitrypsin, to augment the anti-inflammatory potential of the mesenchymal stromal cells. We show that apceth-201 mesenchymal stromal cells efficiently suppress T cell proliferation and polarize macrophages to an anti-inflammatory M2 type, in vitro. To assess the in vivo efficacy of apceth-201, it was tested in two different mouse models of acute graft-versus-host disease. Control animals in a humanized model succumbed quickly to disease, whereas median survival was doubled in apceth-201-treated animals. The product was also tested in a graft-versus-host disease model system that closely mimics haploidentical hematopoietic stem cell transplantation, an approach that is now being evaluated for use in the clinic. Control animals succumbed quickly to disease, whereas treatment with apceth-201 resulted in long-term survival of 57% of the animals. Within 25 days after the second injection, clinical scores returned to baseline in responding animals, indicating complete resolution of graft-versus-host disease. These promising data have led to planning of a phase I study using apceth-201.


Asunto(s)
Expresión Génica , Enfermedad Injerto contra Huésped/etiología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , alfa 1-Antitripsina/genética , Animales , Quimiotaxis de Leucocito/inmunología , Citocinas/metabolismo , Dependovirus/genética , Modelos Animales de Enfermedad , Orden Génico , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Enfermedad Injerto contra Huésped/mortalidad , Enfermedad Injerto contra Huésped/terapia , Xenoinjertos , Mediadores de Inflamación/metabolismo , Activación de Linfocitos/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Trasplante de Células Madre Mesenquimatosas/efectos adversos , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Ratones , Especificidad de Órganos/genética , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Trasplante Homólogo , Resultado del Tratamiento , alfa 1-Antitripsina/metabolismo
6.
Mol Pharm ; 16(6): 2462-2469, 2019 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-31095395

RESUMEN

Delivering peptides and proteins with intracellular function represents a promising avenue for therapeutics, but remains a challenge due to the selective permeability of the plasma membrane. The successful delivery of cytosolically active proteins would enable many opportunities, including improved vaccine development through major histocompatibility complex (MHC) class I antigen display. Extended research using cell-penetrating peptides (CPPs) has aimed to facilitate intracellular delivery of exogenous proteins with some success. A new class of polymer-based mimics termed protein transduction domain mimics (PTDMs), which maintain the positive charge and amphiphilic nature displayed by many CPPs, was developed using a poly-norbornene-based backbone. Herein, we use a previously characterized PTDM to investigate delivery of the model antigen SIINFEKL into leukocytes. Peptide delivery into over 90% of CD14+ monocytes was detected in less than 15 min with nominal inflammatory cytokine response and high cell viability. The co-delivery of a TLR9 agonist and antigen using the PTDM into antigen-presenting cells in vitro showed presentation of SIINFEKL in association with MHC class I molecules, in addition to upregulation of classical differentiation markers revealing the ability of the PTDM to successfully deliver cargo intracellularly and show application in the field of immunotherapy.


Asunto(s)
Monocitos/metabolismo , Presentación de Antígeno/fisiología , Supervivencia Celular/fisiología , Péptidos de Penetración Celular/metabolismo , Células Dendríticas/metabolismo , Citometría de Flujo , Voluntarios Sanos , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Receptores de Lipopolisacáridos/metabolismo , Células THP-1 , Receptor Toll-Like 9/metabolismo
7.
Bioconjug Chem ; 29(8): 2679-2690, 2018 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-30080401

RESUMEN

Delivering proteins into the intracellular environment is a critical step toward probing vital cellular processes for the purposes of ultimately developing new therapeutics. Polymeric carriers are widely used to facilitate protein delivery with guanidinium-rich macromolecules leading the way within this category. Although binding interactions between natural proteins and synthetic polymers have been studied extensively, the relationship between polymer-protein binding and intracellular delivery is seldom explored. Elucidating the role of cargo binding in delivery is a promising direction that is expected to provide new insights that further optimize intracellular protein delivery. Herein, model polymeric carriers called protein transduction domain mimics (PTDMs) were studied for their ability to bind to a variety of protein cargoes, including an antibody, where the proteins encompassed a range of sizes (∼16-151 kDa) and isoelectric points (4.7-11.4). The PTDM-protein complexes were also delivered into Jurkat T cells in an attempt to establish a general correlation between binding ability and delivery outcomes. Binding assays resulted in a vast range of dissociation constants (Kd), which spanned from 3.5 to 4820 nM and indicated a variety of binding strengths between PTDM and protein. More significantly, PTDMs preferentially bound certain types of proteins over others, such as the antibody fragment over the whole antibody. Furthermore, increased PTDM-protein binding affinity did not correlate with protein delivery, suggesting that the successful internalization of complexes is independent of binding equilibrium. Although binding did not correlate with internalization here, the potential for binding affinity to impact other aspects of delivery, like cargo functionality inside the cell, remains an open possibility.


Asunto(s)
Polímeros/química , Proteínas/administración & dosificación , Avidina/química , Citoplasma/metabolismo , Sistemas de Liberación de Medicamentos , Endocitosis , Fluoresceína-5-Isotiocianato/química , Colorantes Fluorescentes/química , Proteínas Fluorescentes Verdes/administración & dosificación , Proteínas Fluorescentes Verdes/metabolismo , Guanidina/química , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Inmunoglobulina G/metabolismo , Células Jurkat , Muramidasa/química , Unión Proteica , Proteínas/metabolismo , Estreptavidina/química
8.
Nat Rev Immunol ; 7(1): 64-75, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17170755

RESUMEN

For many years, researchers have focused on the contribution of Notch signalling to lymphoid development. Only recently have investigators begun to ask what role, if any, Notch has during the activation and differentiation of naive CD4(+) T cells in the periphery. As interest in this issue grows, it is becoming increasingly clear that the main role of Notch signalling, to regulate cell-fate decisions, might also be influential in peripheral T cells.


Asunto(s)
Diferenciación Celular/inmunología , Activación de Linfocitos/inmunología , Receptores Notch/inmunología , Transducción de Señal/inmunología , Linfocitos T/citología , Linfocitos T/inmunología , Animales , Humanos
9.
Blood ; 125(13): 2087-94, 2015 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-25647836

RESUMEN

Aplastic anemia (AA) is a disease characterized by T-cell-mediated destruction of bone marrow (BM) hematopoietic stem and progenitor cells. Physiologically, T cells migrate to the BM in response to chemokines, such as SDF-1α, the ligand for CXCR4. However, how T cells traffic to the BM in AA is poorly understood. CXCR4 is aberrantly expressed in immune-mediated diseases and its regulation by nuclear factor-κB (NF-κB) in cancer models is well documented. In this study, we show that CXCR4 is highly expressed on BM-infiltrating CD4(+) and CD8(+) T cells in a mouse model of AA. Inhibiting CXCR4 in AA mice, using CXCR4(-/-) splenocytes or AMD3100, significantly reduced BM infiltration of T cells. We also report that NF-κB occupancy at the CXCR4 promoter is enhanced in BM-infiltrating CD8(+) T cells of AA mice. Moreover, inhibiting NF-κB signaling in AA mice using Bay11 or dehydroxymethylepoxyquinomicin, or transferring p50(-/-) splenocytes, decreased CXCR4 expression on CD8(+) T cells, significantly reduced BM infiltration of T cells, and strongly attenuated disease symptoms. Remarkably, therapeutic administration of Bay11 significantly extended survival of AA mice. Overall, we demonstrate that CXCR4 mediates migration of pathogenic T cells to the BM in AA mice, and inhibiting NF-κB signaling may represent a novel therapeutic approach to treating AA.


Asunto(s)
Anemia Aplásica , Médula Ósea/inmunología , Linfocitos T CD8-positivos/metabolismo , Quimiotaxis de Leucocito , Modelos Animales de Enfermedad , Ratones , Receptores CXCR4/metabolismo , Anemia Aplásica/genética , Anemia Aplásica/inmunología , Anemia Aplásica/metabolismo , Anemia Aplásica/patología , Animales , Médula Ósea/patología , Linfocitos T CD8-positivos/inmunología , Quimiotaxis de Leucocito/genética , Femenino , Humanos , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/metabolismo , Receptores CXCR4/genética
10.
Chemistry ; 23(28): 6858-6863, 2017 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-28370636

RESUMEN

Cell-penetrating peptides are an important class of molecules with promising applications in bioactive cargo delivery. A diverse series of guanidinium-containing polymeric cell-penetrating peptide mimics (CPPMs) with varying backbone chemistries was synthesized and assessed for delivery of both GFP and fluorescently tagged siRNA. Specifically, we examined CPPMs based on norbornene, methacrylate, and styrene backbones to determine how backbone structure impacted internalization of these two cargoes. Either charge content or degree of polymerization was held constant at 20, with diguanidinium norbornene molecules being polymerized to both 10 and 20 repeat units. Generally, homopolymer CPPMs delivered low amounts of siRNA into Jurkat T cells, with no apparent backbone dependence; however, by adding a short hydrophobic methyl methacrylate block to the guanidinium-rich methacrylate polymer, siRNA delivery to nearly the entire cell population was achieved. Protein internalization yielded similar results for most of the CPPMs, though the block polymer was unable to deliver proteins. In contrast, the styrene-based CPPM yielded the highest internalization for GFP (≈40 % of cells affected), showing that indeed backbone chemistry impacts protein delivery, specifically through the incorporation of an aromatic group. These results demonstrate that an understanding of how polymer structure affects cargo-dependent internalization is critical to designing new, more effective CPPMs.


Asunto(s)
Materiales Biocompatibles/síntesis química , Guanidina/química , Polímeros/química , Materiales Biocompatibles/química , Péptidos de Penetración Celular/química , Citometría de Flujo , Proteínas Fluorescentes Verdes/química , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Células Jurkat , Metilmetacrilato/química , Norbornanos/química , ARN Interferente Pequeño/química , ARN Interferente Pequeño/metabolismo , Transfección/métodos
11.
Mol Ther ; 24(12): 2118-2130, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27633441

RESUMEN

Targeting cellular proteins with antibodies, to better understand cellular signaling pathways in the context of disease modulation, is a fast-growing area of investigation. Humanized antibodies are increasingly gaining attention for their therapeutic potential, but the collection of cellular targets is limited to those secreted from cells or expressed on the cell surface. This approach leaves a wealth of intracellular proteins unexplored as putative targets for antibody binding. Protein kinase Cθ (PKCθ) is essential to T cell activation, proliferation, and differentiation, and its phosphorylation at specific residues is required for its activity. Here we report on the design, synthesis, and characterization of a protein transduction domain mimic capable of efficiently delivering an antibody against phosphorylated PKCθ (Thr538) into human peripheral mononuclear blood cells and altering expression of downstream indicators of T cell activation and differentiation. We used a humanized, lymphocyte transfer model of graft-versus-host disease, to evaluate the durability of protein transduction domain mimic:Anti-pPKCθ modulation, when delivered into human peripheral mononuclear blood cells ex vivo. We demonstrate that protein transduction domain mimic:Antibody complexes can be readily introduced with high efficacy into hard-to-transfect human peripheral mononuclear blood cells, eliciting a biological response sufficient to alter disease progression. Thus, protein transduction domain mimic:Antibody delivery may represent an efficient ex vivo approach to manipulating cellular responses by targeting intracellular proteins.


Asunto(s)
Anticuerpos Monoclonales Humanizados/administración & dosificación , Péptidos de Penetración Celular/síntesis química , Enfermedad Injerto contra Huésped/inmunología , Isoenzimas/antagonistas & inhibidores , Leucocitos Mononucleares/efectos de los fármacos , Proteína Quinasa C/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales Humanizados/química , Anticuerpos Monoclonales Humanizados/farmacología , Diferenciación Celular , Proliferación Celular , Péptidos de Penetración Celular/química , Humanos , Inmunomodulación , Leucocitos Mononucleares/inmunología , Activación de Linfocitos , Ratones , Fosforilación/efectos de los fármacos , Proteína Quinasa C-theta , Transducción de Señal/efectos de los fármacos , Células TH1/inmunología
12.
Biomacromolecules ; 17(10): 3205-3212, 2016 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-27599388

RESUMEN

A fundamental understanding of how polymer structure impacts internalization and delivery of biologically relevant cargoes, particularly small interfering ribonucleic acid (siRNA), is of critical importance to the successful design of improved delivery reagents. Herein we report the use of ring-opening metathesis polymerization (ROMP) methods to synthesize two series of guanidinium-rich protein transduction domain mimics (PTDMs): one based on an imide scaffold that contains one guanidinium moiety per repeat unit, and another based on a diester scaffold that contains two guanidinium moieties per repeat unit. By varying both the degree of polymerization and, in effect, the relative number of cationic charges in each PTDM, the performances of the two ROMP backbones for siRNA internalization were evaluated and compared. Internalization of fluorescently labeled siRNA into Jurkat T cells demonstrated that fluorescein isothiocyanate (FITC)-siRNA internalization had a charge content dependence, with PTDMs containing approximately 40 to 60 cationic charges facilitating the most internalization. Despite this charge content dependence, the imide scaffold yielded much lower viabilities in Jurkat T cells than the corresponding diester PTDMs with similar numbers of cationic charges, suggesting that the diester scaffold is preferred for siRNA internalization and delivery applications. These developments will not only improve our understanding of the structural factors necessary for optimal siRNA internalization, but will also guide the future development of optimized PTDMs for siRNA internalization and delivery.


Asunto(s)
Rastreo Celular , Técnicas de Transferencia de Gen , Polímeros/química , ARN Interferente Pequeño/química , Fluoresceína-5-Isotiocianato/química , Guanidina/química , Humanos , Células Jurkat/efectos de los fármacos , Polímeros/administración & dosificación , Interferencia de ARN/efectos de los fármacos , ARN Interferente Pequeño/administración & dosificación , Transducción Genética
13.
Biomacromolecules ; 17(6): 1969-77, 2016 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-27103189

RESUMEN

Exploring the role of polymer structure for the internalization of biologically relevant cargo, specifically siRNA, is of critical importance to the development of improved delivery reagents. Herein, we report guanidinium-rich protein transduction domain mimics (PTDMs) based on a ring-opening metathesis polymerization scaffold containing tunable hydrophobic moieties that promote siRNA internalization. Structure-activity relationships using Jurkat T cells and HeLa cells were explored to determine how the length of the hydrophobic block and the hydrophobic side chain compositions of these PTDMs impacted siRNA internalization. To explore the hydrophobic block length, two different series of diblock copolymers were synthesized: one series with symmetric block lengths and one with asymmetric block lengths. At similar cationic block lengths, asymmetric and symmetric PTDMs promoted siRNA internalization in the same percentages of the cell population regardless of the hydrophobic block length; however, with 20 repeat units of cationic charge, the asymmetric block length had greater siRNA internalization, highlighting the nontrivial relationships between hydrophobicity and overall cationic charge. To further probe how the hydrophobic side chains impacted siRNA internalization, an additional series of asymmetric PTDMs was synthesized that featured a fixed hydrophobic block length of five repeat units that contained either dimethyl (dMe), methyl phenyl (MePh), or diphenyl (dPh) side chains and varied cationic block lengths. This series was further expanded to incorporate hydrophobic blocks consisting of diethyl (dEt), diisobutyl (diBu), and dicyclohexyl (dCy) based repeat units to better define the hydrophobic window for which our PTDMs had optimal activity. High-performance liquid chromatography retention times quantified the relative hydrophobicities of the noncationic building blocks. PTDMs containing the MePh, diBu, and dPh hydrophobic blocks were shown to have superior siRNA internalization capabilities compared to their more and less hydrophobic counterparts, demonstrating a critical window of relative hydrophobicity for optimal internalization. This better understanding of how hydrophobicity impacts PTDM-induced internalization efficiencies will help guide the development of future delivery reagents.


Asunto(s)
Materiales Biomiméticos , Portadores de Fármacos , Guanidina/química , Polímeros , ARN Interferente Pequeño/administración & dosificación , Materiales Biomiméticos/síntesis química , Materiales Biomiméticos/química , Portadores de Fármacos/síntesis química , Portadores de Fármacos/química , Técnicas de Transferencia de Gen , Células HeLa , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Células Jurkat , Polímeros/síntesis química , Polímeros/química , Dominios Proteicos , Interferencia de ARN/efectos de los fármacos , ARN Interferente Pequeño/genética , Relación Estructura-Actividad , Transducción Genética
14.
Biomacromolecules ; 16(10): 3172-9, 2015 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-26324222

RESUMEN

RNA interference is gaining attention as a means to explore new molecular pathways and for its potential as a therapeutic; however, its application in immortal and primary T cells is limited due to challenges with efficient delivery in these cell types. Herein, we report the development of guanidinium-rich protein transduction domain mimics (PTDMs) based on a ring-opening metathesis polymerization scaffold that delivers siRNA into Jurkat T cells and human peripheral blood mononuclear cells (hPBMCs). Homopolymer and block copolymer PTDMs with varying numbers of guanidinium moieties were designed and tested to assess the effect cationic charge content and the addition of a segregated, hydrophobic block had on siRNA internalization and delivery. Internalization of fluorescently labeled siRNA into Jurkat T cells illustrates that the optimal cationic charge content, 40 charges per polymer, leads to higher efficiencies, with block copolymers outperforming their homopolymer counterparts. PTDMs also outperformed commercial reagents commonly used for siRNA delivery applications. Select PTDM candidates were further screened to assess the role the PTDM structure has on the delivery of biologically active siRNA into primary cells. Specifically, siRNA to hNOTCH1 was delivered to hPBMCs enabling 50-80% knockdown efficiencies, with longer PTDMs showing improved protein reduction. By evaluating the PTDM design parameters for siRNA delivery, more efficient PTDMs were discovered that improved delivery and gene (NOTCH) knockdown in T cells. Given the robust delivery of siRNA by these novel PTDMs, their development should aid in the exploration of T cell molecular pathways leading eventually to new therapeutics.


Asunto(s)
Guanidina/química , Imitación Molecular , Proteínas/química , ARN Interferente Pequeño/administración & dosificación , Linfocitos T/metabolismo , Células HeLa , Humanos , Células Jurkat
15.
Biomacromolecules ; 15(3): 812-20, 2014 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-24506414

RESUMEN

A new series of synthetic protein transduction domain mimics (PTDMs) was designed to analyze the importance of guanidine and phenyl group segregation along the backbone on their membrane interaction and cellular internalization abilities. ROMP was utilized to synthesize three polymers: nonsegregated homopolymers, intermediately segregated gradient copolymers, and strongly segregated block copolymers. In order to understand the role of functional group segregation on activity, it was important to design monomers that enabled these three different polymer topologies, or constitutional macromolecular isomers, to be prepared with identical chemical compositions. The structure-activity relationships were evaluated by both a biophysical assay, using dye-loaded vesicles, and by in vitro cellular uptake studies of fluorescently labeled chains. The results showed that functional group segregation impacts activity. In general, the nonsegregated homopolymer was the most active in both assays but also showed larger, ill-defined aggregates compared to either the gradient or block copolymers. It was also the most cytotoxic of the three isomers. As a result, the gradient copolymer with intermediate segregation optimizes activity and solubility with low cytotoxicity. This study gives new design guidelines for the development of PTDMs.


Asunto(s)
Secuencia de Aminoácidos , Interacciones Hidrofóbicas e Hidrofílicas , Proteínas/química , Relación Estructura-Actividad , Humanos , Sustancias Macromoleculares/química , Metacrilatos/química , Polímeros/química , Proteínas/síntesis química , Solubilidad
16.
Mol Ther ; 21(1): 201-9, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23070119

RESUMEN

RNA interference technology has recently been highlighted as a powerful research method as well as a potential therapeutic treatment for several diseases. However, the delivery of small interfering RNA (siRNA) into T cell lines and primary blood cells is exceedingly challenging, as they are resistant to transfection by conventional reagents. As a result, there is an unmet need for nonviral, efficient, and easily prepared carriers for siRNA delivery into hard-to-transfect cell types. Here, we report a novel system based on protein transduction domain mimics (PTDMs), generated by ring opening metathesis polymerization, for intracellular delivery of siRNA molecules. PTDM-based siRNA delivery induced efficient NOTCH1 knockdown in Jurkat T cells and human peripheral blood mononuclear cells without any measured toxicity. Furthermore, delivering siRNA to NOTCH1 in human peripheral blood cells modulated cell proliferation and differentiation of T cells into T(H)1 cells.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , ARN Interferente Pequeño/genética , Receptor Notch1/genética , Transducción Genética , Linfocitos T CD4-Positivos/citología , Diferenciación Celular , Técnicas de Silenciamiento del Gen , Humanos , Células Jurkat , Interferencia de ARN
17.
Front Immunol ; 15: 1396486, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38694497

RESUMEN

Bone marrow failure (BMF) has become one of the most studied autoimmune disorders, particularly due to its prevalence both as an inherited disease, but also as a result of chemotherapies. BMF is associated with severe symptoms such as bleeding episodes and susceptibility to infections, and often has underlying characteristics, such as anemia, thrombocytopenia, and neutropenia. The current treatment landscape for BMF requires stem cell transplantation or chemotherapies to induce immune suppression. However, there is limited donor cell availability or dose related toxicity associated with these treatments. Optimizing these treatments has become a necessity. Polymer-based materials have become increasingly popular, as current research efforts are focused on synthesizing novel cell matrices for stem cell expansion to solve limited donor cell availability, as well as applying polymer delivery vehicles to intracellularly deliver cargo that can aid in immunosuppression. Here, we discuss the importance and impact of polymer materials to enhance therapeutics in the context of BMF.


Asunto(s)
Polímeros , Humanos , Polímeros/química , Animales , Enfermedades de la Médula Ósea/inducido químicamente , Enfermedades de la Médula Ósea/terapia , Trastornos de Fallo de la Médula Ósea/terapia , Materiales Biocompatibles
18.
Transplant Cell Ther ; 30(1): 79.e1-79.e10, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37924979

RESUMEN

Graft-versus-host disease (GVHD) is a primary and often lethal complication of allogenic hematopoietic stem cell transplantation (HSCT). Prophylactic regimens for GVHD are given as standard pretransplantation therapy; however, up to 50% of these patients develop acute GVHD (aGVHD) and require additional immunosuppressive intervention. Using a mouse GVHD model, we previously showed that injecting mice with exopolysaccharide (EPS) from Bacillus subtilis prior to GVHD induction significantly increased 80-day survival after transplantation of complete allogeneic major histocompatibility complex-mismatched cells. To ask whether EPS might also inhibit GVHD in humans, we used humanized NSG-HLA-A2 mice and induced GVHD by i.v. injection of A2neg human peripheral blood mononuclear cells (PBMCs). Because we could not inject human donors with EPS, we transferred EPS-pretreated dendritic cells (DCs) to inhibit aGVHD. We derived these DCs from CD34+ human cord blood cells, treated them with EPS, and then injected them together with PBMCs into the NSG-HLA-A2 mice. We found that all mice that received untreated DCs were dead by day 35, whereas 25% of mice receiving EPS-treated DCs (EPS-DCs) survived. This DC cell therapy could be readily translatable to humans, because we can generate large numbers of human EPS-DCs and use them as an "off the shelf" treatment for patients undergoing HSCT.


Asunto(s)
Enfermedad Injerto contra Huésped , Antígeno HLA-A2 , Animales , Humanos , Trasplante Homólogo/efectos adversos , Leucocitos Mononucleares , Enfermedad Injerto contra Huésped/prevención & control , Modelos Animales de Enfermedad , Células Dendríticas
19.
Curr Top Microbiol Immunol ; 360: 99-114, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22695917

RESUMEN

For T cells to become fully activated, they must integrate a myriad of signals, both extrinsic and intrinsic. External stimuli accrued through various cell surface receptors are transduced and amplified through a coordinated circuitry of signaling cascades that ultimately result in the transcription of new genes. Along the way, extracellular and intracellular signaling components function to impart a fully activated state. Evidence is accumulating to show that the Notch family of cell surface receptors, long known to function as transcriptional regulators through their interactions with the canonical nuclear binding protein CSL/RBP-J, may also be playing an as-yet-unappreciated role in T cell activation by virtue of its signaling via non-canonical as well as nonnuclear mechanisms. In this review we will discuss these and other better-known means by which Notch signaling influences T cell responses.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Secretasas de la Proteína Precursora del Amiloide/genética , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Linfocitos T CD4-Positivos/citología , Diferenciación Celular , Núcleo Celular/genética , Núcleo Celular/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Humanos , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Activación de Linfocitos/genética , Ratones , Receptores Notch/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
20.
J Immunol ; 187(2): 692-701, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21685328

RESUMEN

Th17 cells are known to play a critical role in adaptive immune responses to several important extracellular pathogens. Additionally, Th17 cells are implicated in the pathogenesis of several autoimmune and inflammatory disorders as well as in cancer. Therefore, it is essential to understand the mechanisms that regulate Th17 differentiation. Notch signaling is known to be important at several stages of T cell development and differentiation. In this study, we report that Notch1 is activated in both mouse and human in vitro-polarized Th17 cells and that blockade of Notch signaling significantly downregulates the production of Th17-associated cytokines, suggesting an intrinsic requirement for Notch during Th17 differentiation in both species. We also present evidence, using promoter reporter assays, knockdown studies, as well as chromatin immunoprecipitation, that IL-17 and retinoic acid-related orphan receptor γt are direct transcriptional targets of Notch signaling in Th17 cells. Finally, in vivo inhibition of Notch signaling reduced IL-17 production and Th17-mediated disease progression in experimental autoimmune encephalomyelitis, a mouse model of multiple sclerosis. Thus, this study highlights the importance of Notch signaling in Th17 differentiation and indicates that selective targeted therapy against Notch may be an important tool to treat autoimmune disorders, including multiple sclerosis.


Asunto(s)
Diferenciación Celular/inmunología , Receptor Notch1/fisiología , Transducción de Señal/inmunología , Células Th17/inmunología , Animales , Células Cultivadas , Citocinas/antagonistas & inhibidores , Citocinas/fisiología , Regulación hacia Abajo/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Encefalomielitis Autoinmune Experimental/terapia , Células HEK293 , Humanos , Interleucina-17/antagonistas & inhibidores , Interleucina-17/metabolismo , Ratones , Ratones Endogámicos C57BL , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/patología , Esclerosis Múltiple/terapia , Células Th17/metabolismo , Células Th17/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA