Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
J Immunol ; 211(11): 1680-1692, 2023 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-37850965

RESUMO

Nucleic acid vaccines, including both RNA and DNA platforms, are key technologies that have considerable promise in combating both infectious disease and cancer. However, little is known about the extrinsic factors that regulate nucleic acid vaccine responses and which may determine their effectiveness. The microbiome is recognized as a significant regulator of immune development and response, whose role in regulating some traditional vaccine platforms has recently been discovered. Using germ-free and specific pathogen-free mouse models in combination with different protein, DNA, and mRNA vaccine regimens, we demonstrate that the microbiome is a significant regulator of nucleic acid vaccine immunogenicity. Although the presence of the microbiome enhances CD8+ T cell responses to mRNA lipid nanoparticle immunization, the microbiome suppresses Ig and CD4+ T cell responses to DNA-prime, DNA-protein-boost immunization, indicating contrasting roles for the microbiome in the regulation of these different nucleic acid vaccine platforms. In the case of mRNA lipid nanoparticle vaccination, germ-free mice display reduced dendritic cell/macrophage activation that may underlie the deficient vaccine response. Our study identifies the microbiome as a relevant determinant of nucleic acid vaccine response with implications for continued therapeutic development and deployment of these vaccines.


Assuntos
Microbiota , Vacinas de DNA , Camundongos , Animais , Vacinas Baseadas em Ácido Nucleico , Linfócitos T CD8-Positivos , DNA , RNA Mensageiro , Imunização Secundária
2.
Sci Transl Med ; 15(686): eabn3464, 2023 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-36867683

RESUMO

As mRNA vaccines have proved to be very successful in battling the coronavirus disease 2019 (COVID-19) pandemic, this new modality has attracted widespread interest for the development of potent vaccines against other infectious diseases and cancer. Cervical cancer caused by persistent human papillomavirus (HPV) infection is a major cause of cancer-related deaths in women, and the development of safe and effective therapeutic strategies is urgently needed. In the present study, we compared the performance of three different mRNA vaccine modalities to target tumors associated with HPV-16 infection in mice. We generated lipid nanoparticle (LNP)-encapsulated self-amplifying mRNA as well as unmodified and nucleoside-modified non-replicating mRNA vaccines encoding a chimeric protein derived from the fusion of the HPV-16 E7 oncoprotein and the herpes simplex virus type 1 glycoprotein D (gDE7). We demonstrated that single low-dose immunizations with any of the three gDE7 mRNA vaccines induced activation of E7-specific CD8+ T cells, generated memory T cell responses capable of preventing tumor relapses, and eradicated subcutaneous tumors at different growth stages. In addition, the gDE7 mRNA-LNP vaccines induced potent tumor protection in two different orthotopic mouse tumor models after administration of a single vaccine dose. Last, comparative studies demonstrated that all three gDE7 mRNA-LNP vaccines proved to be superior to gDE7 DNA and gDE7 recombinant protein vaccines. Collectively, we demonstrated the immunogenicity and therapeutic efficacy of three different mRNA vaccines in extensive comparative experiments. Our data support further evaluation of these mRNA vaccines in clinical trials.


Assuntos
Vacinas Anticâncer , Neoplasias , Infecções por Papillomavirus , Vacinas contra Papillomavirus , Vacinas de DNA , Animais , Feminino , Camundongos , Linfócitos T CD8-Positivos , Modelos Animais de Doenças , Imunização , Camundongos Endogâmicos C57BL , Neoplasias/terapia , Proteínas E7 de Papillomavirus/genética , Infecções por Papillomavirus/complicações , Infecções por Papillomavirus/prevenção & controle , Vacinas contra Papillomavirus/genética , Proteínas Recombinantes , RNA Mensageiro/genética
3.
bioRxiv ; 2023 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-36824851

RESUMO

Nucleic acid vaccines, including both RNA and DNA platforms, are key technologies that have considerable promise in combating both infectious disease and cancer. However, little is known about the extrinsic factors that regulate nucleic acid vaccine responses and which may determine their effectiveness. The microbiome is recognized as a significant regulator of immune development and response, whose role in regulating some traditional vaccine platforms has recently been discovered. Using germ-free and specific-pathogen-free mouse models in combination with different protein, DNA, and mRNA vaccine regimens, we demonstrate that the microbiome is a significant regulator of nucleic acid vaccine immunogenicity. While the presence of the microbiome enhances CD8+ T cell responses to mRNA lipid nanoparticle (LNP) immunization, the microbiome suppresses immunoglobulin and CD4+ T cell responses to DNA-prime, DNA-protein-boost immunization, indicating contrasting roles for the microbiome in the regulation of these different nucleic acid vaccine platforms. In the case of mRNA-LNP vaccination, germ-free mice display reduced dendritic cell/macrophage activation that may underlie the deficient vaccine response. Our study identifies the microbiome as a relevant determinant of nucleic acid vaccine response with implications for their continued therapeutic development and deployment.

4.
Commun Biol ; 6(1): 188, 2023 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-36805684

RESUMO

Herein, we studied the impact of empty LNP (eLNP), component of mRNA-based vaccine, on anti-viral pathways and immune function of cells from young and aged individuals. eLNP induced maturation of monocyte derived dendritic cells (MDDCs). We further show that eLNP upregulated CD40 and induced cytokine production in multiple DC subsets and monocytes. This coincided with phosphorylation of TANK binding kinase 1 (pTBK1) and interferon response factor 7 (pIRF7). In response to eLNP, healthy older adults (>65 yrs) have decreased CD40 expression, and IFN-γ output compared to young adults (<65 yrs). Additionally, cells from older adults have a dysregulated anti-viral signaling response to eLNP stimulation, measured by the defect in type I IFN production, and phagocytosis. Overall, our data show function of eLNP in eliciting DC maturation and innate immune signaling pathways that is impaired in older adults resulting in lower immune responses to SARS-CoV-2 mRNA-based vaccines.


Assuntos
COVID-19 , Adulto Jovem , Humanos , Idoso , SARS-CoV-2 , Células Apresentadoras de Antígenos , Antígenos CD40 , RNA Mensageiro
5.
Int J Mol Sci ; 23(23)2022 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-36498833

RESUMO

Treating immunosuppressive tumors represents a major challenge in cancer therapies. Activation of STING signaling has shown remarkable potential to invigorate the immunologically "cold" tumor microenvironment (TME). However, we have shown that STING is silenced in many human cancers, including pancreatic ductal adenocarcinoma (PDAC) and Merkel cell carcinoma (MCC). In this study, we demonstrated that mRNA-lipid nanoparticle (LNP) technology could be used to efficiently deliver naturally occurring constitutively active STING mutant STINGR284S into these cancer cells to reactivate STING antitumor immunity and trigger robust killing of tumor cells. STING agonists are being actively pursued as cancer immunotherapies. However, traditional STING agonists can induce T cell cytotoxicity, counteracting the desired antitumor immune response. In addition, the antitumor efficacy of traditional STING agonists obligatorily depends on STING expression and does not work in STING-silenced cancers. Importantly, we found that STINGR284S mRNA-LNP does not introduce T cell cytotoxicity. Our studies demonstrated that mRNA-LNP delivery of STINGR284S can reactivate the antitumor response without introducing antiproliferative effects in lymphocytic immune cells, overcoming the toxicity and limitations of conventional STING agonists. Our work therefore identifies a novel therapeutic tool for reactivating antitumor immunity in an array of STING-silenced immunologically "cold" tumors that are refractory to current therapies.


Assuntos
Carcinoma de Célula de Merkel , Nanopartículas , Neoplasias Cutâneas , Humanos , RNA Mensageiro/genética , Proteínas de Membrana/metabolismo , Microambiente Tumoral , Imunoterapia
6.
Front Immunol ; 13: 983000, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36311701

RESUMO

An mRNA with unmodified nucleosides induces type I interferons (IFN-I) through the stimulation of innate immune sensors. Whether IFN-I induced by mRNA vaccine is crucial for anti-tumor immune response remains to be elucidated. In this study, we investigated the immunogenicity and anti-tumor responses of mRNA encoding tumor antigens with different degrees of N1-methylpseudouridine (m1Ψ) modification in B16 melanoma model. Our results demonstrated that ovalbumin (OVA) encoding mRNA formulated in a lipid nanoparticle (OVA-LNP) induced substantial IFN-I production and the maturation of dendritic cells (DCs) with negative correlation with increasing percentages of m1Ψ modification. In B16-OVA murine melanoma model, unmodified OVA-LNP significantly reduced tumor growth and prolonged survival, compared to OVA-LNP with m1Ψ modification. This robust anti-tumor effect correlated with the increase in intratumoral CD40+ DCs and the frequency of granzyme B+/IFN-γ+/TNF-α+ polyfunctional OVA peptide-specific CD8+ T cells. Blocking type I IFN receptor completely reversed the anti-tumor immunity of unmodified mRNA-OVA reflected in a significant decrease in OVA-specific IFN-γ secreting T cells and enrichment of PD-1+ tumor-infiltrating T cells. The robust anti-tumor effect of unmodified OVA-LNP was also observed in the lung metastatic tumor model. Finally, this mRNA vaccine was tested using B16 melanoma neoantigens (Pbk-Actn4) which resulted in delayed tumor growth. Taken together, our findings demonstrated that an unmodified mRNA vaccine induces IFN-I production or the downstream signaling cascades which plays a crucial role in inducing robust anti-tumor T cell response for controlling tumor growth and metastasis.


Assuntos
Interferon Tipo I , Melanoma Experimental , Camundongos , Animais , Linfócitos T CD8-Positivos , Uridina , Antígenos de Neoplasias , Ovalbumina , RNA Mensageiro/genética , Vacinas de mRNA
7.
Mol Ther ; 30(9): 2909-2922, 2022 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-35581938

RESUMO

Persistence of chronic hepatitis B (CHB) is attributed to maintenance of the intrahepatic pool of the viral covalently closed circular DNA (cccDNA), which serves as the transcriptional template for all viral gene products required for replication. Current nucleos(t)ide therapies for CHB prevent virus production and spread but have no direct impact on cccDNA or expression of viral genes. We describe a potential curative approach using a highly specific engineered ARCUS nuclease (ARCUS-POL) targeting the hepatitis B virus (HBV) genome. Transient ARCUS-POL expression in HBV-infected primary human hepatocytes produced substantial reductions in both cccDNA and hepatitis B surface antigen (HBsAg). To evaluate ARCUS-POL in vivo, we developed episomal adeno-associated virus (AAV) mouse and non-human primate (NHP) models containing a portion of the HBV genome serving as a surrogate for cccDNA. Clinically relevant delivery was achieved through systemic administration of lipid nanoparticles containing ARCUS-POL mRNA. In both mouse and NHP, we observed a significant decrease in total AAV copy number and high on-target indel frequency. In the case of the mouse model, which supports HBsAg expression, circulating surface antigen was durably reduced by 96%. Together, these data support a gene-editing approach for elimination of cccDNA toward an HBV cure.


Assuntos
Hepatite B Crônica , Hepatite B , Animais , Antivirais , DNA Circular/genética , DNA Viral/genética , Dependovirus/genética , Hepatite B/terapia , Antígenos de Superfície da Hepatite B/genética , Antígenos de Superfície da Hepatite B/uso terapêutico , Vírus da Hepatite B/genética , Humanos , Lipossomos , Camundongos , Nanopartículas , Replicação Viral
8.
Pharmaceutics ; 14(2)2022 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-35214130

RESUMO

The world raced to develop vaccines to protect against the rapid spread of SARS-CoV-2 infection upon the recognition of COVID-19 as a global pandemic. A broad spectrum of candidates was evaluated, with mRNA-based vaccines emerging as leaders due to how quickly they were available for emergency use while providing a high level of efficacy. As a modular technology, the mRNA-based vaccines benefitted from decades of advancements in both mRNA and delivery technology prior to the current global pandemic. The fundamental lessons of the utility of mRNA as a therapeutic were pioneered by Dr. Katalin Kariko and her colleagues, perhaps most notably in collaboration with Drew Weissman at University of Pennsylvania, and this foundational work paved the way for the development of the first ever mRNA-based therapeutic authorized for human use, COMIRNATY®. In this Special Issue of Pharmaceutics, we will be honoring Dr. Kariko for her great contributions to the mRNA technology to treat diseases with unmet needs. In this review article, we will focus on the delivery platform, the lipid nanoparticle (LNP) carrier, which allowed the potential of mRNA therapeutics to be realized. Similar to the mRNA technology, the development of LNP systems has been ongoing for decades before culminating in the success of the first clinically approved siRNA-LNP product, ONPATTRO®, a treatment for an otherwise fatal genetic disease called transthyretin amyloidosis. Lessons learned from the siRNA-LNP experience enabled the translation into the mRNA platform with the eventual authorization and approval of the mRNA-LNP vaccines against COVID-19. This marks the beginning of mRNA-LNP as a pharmaceutical option to treat genetic diseases.

9.
Immunity ; 54(12): 2877-2892.e7, 2021 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-34852217

RESUMO

Adjuvants are critical for improving the quality and magnitude of adaptive immune responses to vaccination. Lipid nanoparticle (LNP)-encapsulated nucleoside-modified mRNA vaccines have shown great efficacy against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), but the mechanism of action of this vaccine platform is not well-characterized. Using influenza virus and SARS-CoV-2 mRNA and protein subunit vaccines, we demonstrated that our LNP formulation has intrinsic adjuvant activity that promotes induction of strong T follicular helper cell, germinal center B cell, long-lived plasma cell, and memory B cell responses that are associated with durable and protective antibodies in mice. Comparative experiments demonstrated that this LNP formulation outperformed a widely used MF59-like adjuvant, AddaVax. The adjuvant activity of the LNP relies on the ionizable lipid component and on IL-6 cytokine induction but not on MyD88- or MAVS-dependent sensing of LNPs. Our study identified LNPs as a versatile adjuvant that enhances the efficacy of traditional and next-generation vaccine platforms.


Assuntos
Linfócitos B/imunologia , Vacinas contra COVID-19/imunologia , COVID-19/imunologia , Centro Germinativo/imunologia , SARS-CoV-2/fisiologia , Linfócitos T Auxiliares-Indutores/imunologia , Vacinas de mRNA/imunologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Adjuvantes Imunológicos , Animais , Células HEK293 , Humanos , Imunidade Humoral , Interleucina-6/genética , Interleucina-6/metabolismo , Lipossomos/administração & dosagem , Camundongos , Camundongos Endogâmicos BALB C , Nanopartículas/administração & dosagem , Subunidades Proteicas/genética , Vacinas de mRNA/genética
10.
Mol Ther ; 29(9): 2769-2781, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-33992803

RESUMO

It is well established that memory CD8 T cells protect susceptible strains of mice from mousepox, a lethal viral disease caused by ectromelia virus (ECTV), the murine counterpart to human variola virus. While mRNA vaccines induce protective antibody (Ab) responses, it is unknown whether they also induce protective memory CD8 T cells. We now show that immunization with different doses of unmodified or N(1)-methylpseudouridine-modified mRNA (modified mRNA) in lipid nanoparticles (LNP) encoding the ECTV gene EVM158 induced similarly strong CD8 T cell responses to the epitope TSYKFESV, albeit unmodified mRNA-LNP had adverse effects at the inoculation site. A single immunization with 10 µg modified mRNA-LNP protected most susceptible mice from mousepox, and booster vaccination increased the memory CD8 T cell pool, providing full protection. Moreover, modified mRNA-LNP encoding TSYKFESV appended to green fluorescent protein (GFP) protected against wild-type ECTV infection while lymphocytic choriomeningitis virus glycoprotein (GP) modified mRNA-LNP protected against ECTV expressing GP epitopes. Thus, modified mRNA-LNP can be used to create protective CD8 T cell-based vaccines against viral infections.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Vírus da Ectromelia/imunologia , Ectromelia Infecciosa/prevenção & controle , Proteínas Virais/genética , Vacinas de mRNA/administração & dosagem , Animais , Composição de Medicamentos , Ectromelia Infecciosa/imunologia , Imunização Secundária , Memória Imunológica , Lipossomos , Masculino , Camundongos , Nanopartículas , Peptídeos/química , Peptídeos/genética , Peptídeos/imunologia , Pseudouridina/análogos & derivados , Pseudouridina/química , Proteínas Virais/química , Proteínas Virais/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/química , Vacinas Virais/farmacologia , Vacinas de mRNA/química , Vacinas de mRNA/farmacologia
11.
Nat Biomed Eng ; 5(2): 179-189, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33495639

RESUMO

Base editors are RNA-programmable deaminases that enable precise single-base conversions in genomic DNA. However, off-target activity is a concern in the potential use of base editors to treat genetic diseases. Here, we report unbiased analyses of transcriptome-wide and genome-wide off-target modifications effected by cytidine base editors in the liver of mice with phenylketonuria. The intravenous delivery of intein-split cytidine base editors by dual adeno-associated viruses led to the repair of the disease-causing mutation without generating off-target mutations in the RNA and DNA of the hepatocytes. Moreover, the transient expression of a cytidine base editor mRNA and a relevant single-guide RNA intravenously delivered by lipid nanoparticles led to ~21% on-target editing and to the reversal of the disease phenotype; there were also no detectable transcriptome-wide and genome-wide off-target edits. Our findings support the feasibility of therapeutic cytidine base editing to treat genetic liver diseases.


Assuntos
Citidina/genética , DNA/genética , Edição de Genes/métodos , Hepatócitos/metabolismo , RNA/genética , Adenoviridae/fisiologia , Animais , Vetores Genéticos/fisiologia , Células HEK293 , Humanos , Camundongos Endogâmicos C57BL
12.
Mol Ther ; 27(4): 866-877, 2019 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-30902585

RESUMO

It has previously been shown that engineered zinc finger nucleases (ZFNs) can be packaged into adeno-associated viruses (AAVs) and delivered intravenously into mice, non-human primates, and most recently, humans to induce highly efficient therapeutic genome editing in the liver. Lipid nanoparticles (LNPs) are synthetic delivery vehicles that enable repeat administration and are not limited by the presence of preexisting neutralizing antibodies in patients. Here, we show that mRNA encoding ZFNs formulated into LNP can enable >90% knockout of gene expression in mice by targeting the TTR or PCSK9 gene, at mRNA doses 10-fold lower than has ever been reported. Additionally, co-delivering mRNA-LNP containing ZFNs targeted to intron 1 of the ALB locus with AAV packaged with a promoterless human IDS or FIX therapeutic transgene can result in high levels of targeted integration and subsequent therapeutically relevant levels of protein expression in mice. Finally, we show repeat administration of ZFN mRNA-LNP after a single AAV donor dose results in significantly increased levels of genome editing and transgene expression compared to a single dose. These results demonstrate LNP-mediated ZFN mRNA delivery can drive highly efficient levels of in vivo genome editing and can potentially offer a new treatment modality for a variety of diseases.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Edição de Genes/métodos , Nanopartículas/administração & dosagem , RNA Mensageiro/administração & dosagem , Nucleases de Dedos de Zinco/administração & dosagem , Animais , Células Cultivadas , Dependovirus/genética , Feminino , Técnicas de Inativação de Genes , Vetores Genéticos , Hepatócitos/metabolismo , Íntrons/genética , Lipídeos/química , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pré-Albumina/genética , Pró-Proteína Convertase 9/genética , RNA Mensageiro/genética , Transgenes/genética , Nucleases de Dedos de Zinco/farmacologia
13.
Clin Cancer Res ; 21(21): 4845-55, 2015 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-26106075

RESUMO

PURPOSE: Lipid nanoparticle (LNP) formulations facilitate tumor uptake and intracellular processing through an enhanced permeation and retention effect (EPR), and currently multiple products are undergoing clinical evaluation. Clusterin (CLU) is a cytoprotective chaperone induced by androgen receptor (AR) pathway inhibition to facilitate adaptive survival pathway signaling and treatment resistance. In our study, we investigated the efficacy of siRNA tumor delivery using LNP systems in an enzalutamide-resistant (ENZ-R) castration-resistant prostate cancer (CRPC) model. EXPERIMENTAL DESIGN: Gene silencing of a luciferase reporter gene in the PC-3M-luc stable cell line was first assessed in subcutaneous and metastatic PC-3 xenograft tumors. Upon validation, the effect of LNP siRNA targeting CLU in combination with AR antisense oligonucleotides (ASO) was assessed in ENZ-R CRPC LNCaP in vitro and in vivo models. RESULTS: LNP LUC-siRNA silenced luciferase expression in PC-3M-luc subcutaneous xenograft and metastatic models. LNP CLU-siRNA potently suppressed CLU and AR ASO-induced CLU and AKT and ERK phosphorylation in ENZ-R LNCaP cells in vitro, more potently inhibiting ENZ-R cell growth rates and increased apoptosis when compared with AR-ASO monotherapy. In subcutaneous ENZ-R LNCaP xenografts, combinatory treatment of LNP CLU-siRNA plus AR-ASO significantly suppressed tumor growth and serum PSA levels compared with LNP LUC-siRNA (control) and AR-ASO. CONCLUSIONS: LNP siRNA can silence target genes in vivo and enable inhibition of traditionally non-druggable genes like CLU and other promising cotargeting approaches in ENZ-R CRPC therapeutics.


Assuntos
Clusterina/genética , Inativação Gênica , Lipídeos , Nanopartículas , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , RNA Interferente Pequeno/genética , Receptores Androgênicos/genética , Animais , Antineoplásicos/farmacologia , Apoptose/genética , Benzamidas , Linhagem Celular Tumoral , Proliferação de Células , Modelos Animais de Doenças , Progressão da Doença , Resistencia a Medicamentos Antineoplásicos , Expressão Gênica , Genes Reporter , Humanos , Masculino , Camundongos , Imagem Molecular/métodos , Metástase Neoplásica , Nitrilas , Oligonucleotídeos Antissenso/genética , Feniltioidantoína/análogos & derivados , Feniltioidantoína/farmacologia , Neoplasias de Próstata Resistentes à Castração/genética , Neoplasias de Próstata Resistentes à Castração/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Nanomedicine ; 9(2): 233-46, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22698807

RESUMO

The in vivo gene silencing potencies of lipid nanoparticle (LNP)-siRNA systems containing the ionizable cationic lipids DLinDAP, DLinDMA, DLinKDMA, or DLinKC2-DMA can differ by three orders of magnitude. In this study, we examine the uptake and intracellular processing of LNP-siRNA systems containing these cationic lipids in a macrophage cell-line in an attempt to understand the reasons for different potencies. Although uptake of LNP is not dramatically influenced by cationic lipid composition, subsequent processing events can be strongly dependent on cationic lipid species. In particular, the low potency of LNP containing DLinDAP can be attributed to hydrolysis by endogenous lipases following uptake. LNP containing DLinKC2-DMA, DLinKDMA, or DLinDMA, which lack ester linkages, are not vulnerable to lipase digestion and facilitate much more potent gene silencing. The superior potency of DLinKC2-DMA compared with DLinKDMA or DLinDMA can be attributed to higher uptake and improved ability to stimulate siRNA release from endosomes subsequent to uptake. FROM THE CLINICAL EDITOR: This study reports on the in vivo gene silencing potency of lipid nanoparticle-siRNA systems containing ionizable cationic lipids. It is concluded that the superior potency of DLinKC2-DMA compared with DLinKDMA or DLinDMA can be attributed to their higher uptake thus improved ability to stimulate siRNA release from endosome.


Assuntos
Lipídeos/química , Macrófagos/metabolismo , Nanopartículas/química , RNA Interferente Pequeno/administração & dosagem , Animais , Cátions/química , Cátions/metabolismo , Linhagem Celular , Clatrina/metabolismo , Endocitose , Lipase/metabolismo , Metabolismo dos Lipídeos , Camundongos , Pinocitose , Interferência de RNA , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , RNA Interferente Pequeno/farmacocinética , Ribonucleases/metabolismo
15.
Nanomedicine ; 9(5): 665-74, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23219877

RESUMO

Gene silencing activity of lipid nanoparticle (LNP) formulations of siRNA requires LNP surface factors promoting cellular uptake. This study aimed to identify small molecules that enhance cellular uptake of LNP siRNA systems, then use them as LNP-associated ligands to improve gene silencing potency. Screening the Canadian Chemical Biology Network molecules for effects on LNP uptake into HeLa cells found that cardiac glycosides like ouabain and strophanthidin caused the highest uptake. Cardiac glycosides stimulate endocytosis on binding to plasma membrane Na(+)/K(+) ATPase found in all mammalian cells, offering the potential to stimulate LNP uptake into various cell types. A PEG-lipid containing strophanthidin at the end of PEG (STR-PEG-lipid) was synthesized and incorporated into LNP. Compared to non-liganded systems, STR-PEG-lipid enhanced LNP uptake in various cell types. Furthermore, this enhanced uptake improved marker gene silencing in vitro. Addition of STR-PEG-lipid to LNP siRNA may have general utility for enhancing gene silencing potency. FROM THE CLINICAL EDITOR: In this study, the authors identified small molecules that enhance cellular uptake of lipid nanoparticle siRNA systems, then used them as LNP-associated ligands to improve gene silencing potency.


Assuntos
Lipídeos/administração & dosagem , Nanopartículas/administração & dosagem , RNA Interferente Pequeno/genética , Estrofantidina/administração & dosagem , Animais , Endocitose/genética , Inativação Gênica/efeitos dos fármacos , Técnicas de Transferência de Genes , Células HeLa , Humanos , Ligantes , Lipídeos/química , Nanopartículas/química , RNA Interferente Pequeno/química , ATPase Trocadora de Sódio-Potássio/genética , ATPase Trocadora de Sódio-Potássio/metabolismo , Estrofantidina/química
16.
Oncol Rep ; 27(2): 311-7, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22076128

RESUMO

Na+/H+ exchangers (NHEs) are a group of secondary active antiporters that regulate cellular pH, cell volume and ion homeostasis. In humans, nine isoforms (NHE1-NHE9) were identified and characterized as functional NHEs. While a growing body of evidence indicates that NHE1 generates an acidic tumor environment and thereby contributes to tumor invasion, little is known about the role of other NHE isoforms in tumor progression. NHE7 is a unique member of the NHE gene family that dynamically shuttles between the trans-Golgi network, endosomes and the plasma membrane, and regulates the luminal pH of these organelles. Here we show that NHE7-overexpression in breast cancer MDA-MB-231 cells enhances cell overlay, cell-cell adhesion, invasion, anchorage-independent tumor growth and tumor formation in vivo. In contrast, NHE1-overexpression enhances tumor invasion, but it has little effect on cell adhesion or anchorage-independent tumor growth. Pathological examinations of the tumor samples derived from NHE7-overexpressing cells showed a similar appearance to aggressive tumors. Together, these results suggest that NHE7 enhances tumor progression. This is the first report to show the involvement of an organellar NHE in oncogenic processes.


Assuntos
Neoplasias da Mama/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , Adesão Celular/genética , Linhagem Celular Tumoral , Proliferação de Células , Modelos Animais de Doenças , Feminino , Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Camundongos Nus , Invasividade Neoplásica/genética , Trocador 1 de Sódio-Hidrogênio , Trocadores de Sódio-Hidrogênio/genética , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Int J Cancer ; 131(5): E781-90, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22095615

RESUMO

The androgen receptor (AR) plays a critical role in the progression of prostate cancer. Silencing this protein using short-hairpin RNA (shRNA) has been correlated with tumor growth inhibition and decreases in serum prostate specific antigen (PSA). In our study, we have investigated the ability of lipid nanoparticle (LNP) formulations of small-interfering RNA (siRNA) to silence AR in human prostate tumor cell lines in vitro and in LNCaP xenograft tumors following intravenous (i.v.) injection. In vitro screening studies using a panel of cationic lipids showed that LNPs containing the ionizable cationic lipid 2,2-dilinoleyl-4-(2-dimethylaminoethyl)-[1,3]-dioxolane (DLin-KC2-DMA) exhibited the most potent AR silencing effects in LNCaP cells. This is attributed to an optimized ability of DLin-KC2-DMA-containing LNP to be taken up into cells and to release the siRNA into the cell cytoplasm following endocytotic uptake. DLin-KC2-DMA LNPs were also effective in silencing the AR in a wild-type AR expressing cell line, LAPC-4, and a variant AR expressing cell line, CWR22Rv1. Importantly, it is demonstrated that LNP AR-siRNA systems containing DLin-KC2-DMA can silence AR gene expression in distal LNCaP xenograft tumors and decrease serum PSA levels following i.v. injection. To our knowledge, this is the first report demonstrating the feasibility of LNP delivery of siRNA for silencing AR gene expression in vivo.


Assuntos
Antagonistas de Receptores de Andrógenos/farmacologia , Lipídeos , Nanopartículas , Neoplasias da Próstata/genética , Neoplasias da Próstata/terapia , RNA Interferente Pequeno/genética , Receptores Androgênicos/química , Progressão da Doença , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Neoplasias da Próstata/metabolismo , Receptores Androgênicos/genética , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Cell Signal ; 19(5): 978-88, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17207967

RESUMO

NHE7 was identified as the first mammalian organelle-membrane type (Na+, K+)/H+ exchanger that may contribute to the ion homeostasis in the trans-Golgi network (TGN) and endosomes. Here we show that caveolins directly bind to the C-terminal extension of NHE7 by an unconventional binding-module. NHE7 is partly associated with caveolae/lipid raft fractions, and heterologous expression of caveolin dominant-negative mutants as well as cholesterol depriving drugs diminished such associations. In contrast to the wild type NHE7, a deletion mutant lacking the C-terminal extension was predominantly detected in non-caveolae/lipid rafts. We further show that a small fraction of NHE7 is targeted to the cell surface and subsequently internalized. Endocytosis of NHE7 was efficiently inhibited by pharmacological maneuvers that block clathrin-dependent endocytosis, whereas dominant-negative caveolin mutants or methyl beta-cyclodextrin did not affect NHE7-internalization. Thus, NHE7 associates with both caveolae/lipid rafts and non-caveolae/lipid raft, and the two pools likely exhibit separate dynamics.


Assuntos
Caveolinas/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , Animais , Células CHO , Cavéolas/metabolismo , Caveolinas/genética , Linhagem Celular Tumoral , Clatrina/metabolismo , Cricetinae , Cricetulus , Citosol/química , Humanos , Microdomínios da Membrana/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Trocadores de Sódio-Hidrogênio/química , Rede trans-Golgi/metabolismo
19.
Cell Signal ; 19(1): 194-203, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16920332

RESUMO

Na+/H+ exchanger isoform 5 (NHE5) is a brain-enriched NHE that may play important roles in ion homeostasis and cell-volume regulation. However, the regulation mechanism of NHE5 has not been fully elucidated. Here, we show that Receptor for Activated C-kinase 1 (RACK1) directly binds to NHE5 and positively regulates the transporter function. NHE5 co-localized with RACK1 as well as beta1 integrin, paxillin and vinculin, suggesting that NHE5 associates with focal adhesions. By using RACK1 dominant-negative mutants and siRNA, we further show that RACK1 regulates NHE5 both directly and through an integrin-dependent pathway. The NHE5-RACK1 interaction, but not the RACK1-beta1 integrin interaction, was reinforced when cells were spread on an integrin-substrate fibronectin. We propose that RACK1 activates NHE5 both by integrin-dependent and independent pathways, which may coordinate cellular ion homeostasis during cell-matrix adhesion.


Assuntos
Adesões Focais , Proteínas de Ligação ao GTP/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Receptores de Superfície Celular/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , Animais , Células CHO , Adesão Celular , Cricetinae , Cricetulus , Fibronectinas/metabolismo , Proteínas de Ligação ao GTP/genética , Humanos , Integrina beta1/metabolismo , Proteínas de Membrana/genética , Mutação , Proteínas de Neoplasias/genética , Paxilina/metabolismo , Ligação Proteica , Receptores de Quinase C Ativada , Receptores de Superfície Celular/genética , Transdução de Sinais , Trocadores de Sódio-Hidrogênio/genética , Técnicas do Sistema de Duplo-Híbrido , Vinculina/metabolismo
20.
J Cell Sci ; 118(Pt 9): 1885-97, 2005 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-15840657

RESUMO

The mammalian (Na(+),K(+))/H(+) exchanger NHE7 resides chiefly in the trans-Golgi network (TGN) and post-Golgi vesicles where it is thought to contribute to organellar pH homeostasis. However, the mechanisms that underlie the targeting and regulation of NHE7 are unknown. To gain insight into these processes, yeast two-hybrid methodology was used to screen a human brain cDNA library for proteins that interact with the cytoplasmic C-terminus of NHE7. One binding partner we identified was SCAMP2, a member of the secretory carrier membrane protein (SCAMP) gene family. Direct association of these two proteins was further supported by co-immunolocalization and co-immunoprecipitation analyses using transfected cells, by their co-sedimentation in membrane fractions resolved on sucrose density gradients, and by in vitro protein binding assays. Other members of the SCAMP family, such as SCAMP1 and SCAMP5, also associated with NHE7. The majority of the NHE7-SCAMP complexes accumulated at the TGN, but a minor fraction also resided in recycling vesicles. Biochemical analyses indicated that the C-terminal cytoplasmic tail of NHE7 bound preferentially to a highly conserved cytoplasmic loop between the second and the third transmembrane segments (TM2-TM3 loop) of SCAMP2. A deletion mutant of SCAMP2 lacking this region (SCAMP2/Delta184-208) bound weakly to NHE7, but caused a significant fraction of NHE7 and wild-type SCAMP2 to redistribute to a pool of scattered recycling vesicles without noticeably affecting the location of other resident TGN (syntaxin 6) or Golgi cisternae (GM130) proteins. Conversely, a GFP-tagged TM2-TM3 construct of SCAMP2 interacted with NHE7, but also led to the redistribution of NHE7 to dispersed vesicular structures. We propose a model wherein SCAMPs participate in the shuttling of NHE7 between recycling vesicles and the TGN.


Assuntos
Antiporters/fisiologia , Membrana Celular/metabolismo , Proteínas de Membrana Transportadoras/fisiologia , Animais , Antiporters/metabolismo , Autoantígenos , Transporte Biológico , Encéfalo/metabolismo , Células CHO , Proteínas de Transporte/metabolismo , Centrifugação com Gradiente de Concentração , Clonagem Molecular , Cricetinae , Citoplasma/metabolismo , DNA Complementar/metabolismo , Relação Dose-Resposta a Droga , Deleção de Genes , Biblioteca Gênica , Glutationa Transferase/metabolismo , Complexo de Golgi/metabolismo , Proteínas de Fluorescência Verde/química , Proteínas de Fluorescência Verde/metabolismo , Humanos , Imunoprecipitação , Proteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Microscopia de Fluorescência , Modelos Biológicos , Células PC12 , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Qa-SNARE , Ratos , Proteínas Recombinantes de Fusão/química , Trocadores de Sódio-Hidrogênio , Sacarose/farmacologia , Transfecção , Técnicas do Sistema de Duplo-Híbrido
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA