Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
1.
Small ; 19(37): e2300527, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37226374

RESUMO

In this study, extracellular vesicles (EVs) are reimagined as more than just a cellular waste disposal system and are repurposed for cancer immunotherapy. Potent oncolytic EVs (bRSVF-EVs) loaded with misfolded proteins (MPs) are engineered, which are typically considered cellular debris. By impairing lysosomal function using bafilomycin A1 and expressing the respiratory syncytial virus F protein, a viral fusogen, MPs are successfully loaded into the EVs expressing RSVF. bRSVF-EVs preferentially transplant a xenogeneic antigen onto cancer cell membranes in a nucleolin-dependent manner, triggering an innate immune response. Furthermore, bRSVF-EV-mediated direct delivery of MPs into the cancer cell cytoplasm initiates endoplasmic reticulum stress and immunogenic cell death (ICD). This mechanism of action leads to substantial antitumor immune responses in murine tumor models. Importantly, when combined with PD-1 blockade, bRSVF-EV treatment elicits robust antitumor immunity, resulting in prolonged survival and complete remission in some cases. Overall, the findings demonstrate that utilizing tumor-targeting oncolytic EVs for direct cytoplasmic delivery of MPs to induce ICD in cancer cells represents a promising approach for enhancing durable antitumor immunity.


Assuntos
Vesículas Extracelulares , Neoplasias , Camundongos , Animais , Vesículas Extracelulares/metabolismo , Neoplasias/patologia , Citoplasma , Citosol , Imunoterapia/métodos
2.
Small ; 18(15): e2200060, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35229462

RESUMO

Macrophages (Mφs) are characterized by remarkable plasticity, an essential component of chronic inflammation. Thus, an appropriate and timely transition from proinflammatory (M1) to anti-inflammatory (M2) Mφs during wound healing is vital to promoting resolution of acute inflammation and enhancing tissue repair. Herein, exosomes derived from M2-Mφs (M2-Exos), which contain putative key regulators driving Mφ polarization, are used as local microenvironmental cues to induce reprogramming of M1-Mφs toward M2-Mφs for effective wound management. As an injectable controlled release depot for exosomes, hydrolytically degradable poly(ethylene glycol) (PEG) hydrogels (Exogels) are designed and employed for encapsulating M2-Exos to maximize their therapeutic effects in cutaneous wound healing. The degradation time of the hydrogels is adjustable from 6 days or up to 27 days by controlling the crosslinking density and tightness. The localization of M2-Exos leads to a successful local transition from M1-Mφs to M2-Mφs within the lesion for more than 6 days, followed by enhanced therapeutic effects including rapid wound closure and increased healing quality in an animal model for cutaneous wound healing. Collectively, the hydrolytically degradable PEG hydrogel-based exosome delivery system may serve as a potential tool in regulating local polarization state of Mφs, which is crucial for tissue homeostasis and wound repair.


Assuntos
Exossomos , MicroRNAs , Animais , Materiais Biocompatíveis/metabolismo , Preparações de Ação Retardada , Exossomos/metabolismo , Hidrogéis , Inflamação/metabolismo , Macrófagos/metabolismo , MicroRNAs/metabolismo , Cicatrização/fisiologia
3.
Int J Mol Sci ; 22(18)2021 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-34576180

RESUMO

A variety of innate immune cells such as macrophages, dendritic cells, myeloid-derived suppressor cells, natural killer cells, and neutrophils in the tumor microenvironments, contribute to tumor progression. However, while several recent reports have studied the use of immune checkpoint-based cancer immunotherapy, little work has focused on modulating the innate immune cells. This review focuses on the recent studies and challenges of using nanoparticles to target innate immune cells. In particular, we also examine the immunosuppressive properties of certain innate immune cells that limit clinical benefits. Understanding the cross-talk between tumors and innate immune cells could contribute to the development of strategies for manipulating the nanoparticles targeting tumor microenvironments.


Assuntos
Microambiente Tumoral/fisiologia , Animais , Humanos , Imunidade Inata/genética , Imunidade Inata/fisiologia , Células Supressoras Mieloides/metabolismo , Nanopartículas/química , Microambiente Tumoral/genética
4.
Int J Mol Sci ; 22(11)2021 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-34067503

RESUMO

Extracellular vesicles (EV) deliver cargoes such as nucleic acids, proteins, and lipids between cells and serve as an intercellular communicator. As it is revealed that most of the functions associated to EVs are closely related to the immune response, the important role of EVs in inflammatory diseases is emerging. EVs can be functionalized through EV surface engineering and endow targeting moiety that allows for the target specificity for therapeutic applications in inflammatory diseases. Moreover, engineered EVs are considered as promising nanoparticles to develop personalized therapeutic carriers. In this review, we highlight the role of EVs in various inflammatory diseases, the application of EV as anti-inflammatory therapeutics, and the current state of the art in EV engineering techniques.


Assuntos
Vesículas Extracelulares/efeitos dos fármacos , Vesículas Extracelulares/metabolismo , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Animais , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Humanos , Nanopartículas/química
5.
Int J Mol Sci ; 22(1)2020 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-33374978

RESUMO

Exosomes are cell-secreted nanovesicles that naturally contain biomolecular cargoes such as lipids, proteins, and nucleic acids. Exosomes mediate intercellular communication, enabling the transfer biological signals from the donor cells to the recipient cells. Recently, exosomes are emerging as promising drug delivery vehicles due to their strong stability in blood circulation, high biocompatibility, low immunogenicity, and natural targeting ability. In particular, exosomes derived from specific types of cells can carry endogenous signaling molecules with therapeutic potential for cancer treatment, thus presenting a significant impact on targeted drug delivery and therapy. Furthermore, exosomes can be engineered to display targeting moieties on their surface or to load additional therapeutic agents. Therefore, a comprehensive understanding of exosome biogenesis and the development of efficient exosome engineering techniques will provide new avenues to establish convincing clinical therapeutic strategies based on exosomes. This review focuses on the therapeutic applications of exosomes derived from various cells and the exosome engineering technologies that enable the accurate delivery of various types of cargoes to target cells for cancer therapy.


Assuntos
Antineoplásicos/administração & dosagem , Micropartículas Derivadas de Células/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Exossomos/metabolismo , Nanopartículas/metabolismo , Neoplasias/tratamento farmacológico , Animais , Portadores de Fármacos/metabolismo , Humanos , Terapia de Alvo Molecular/métodos
6.
Proc Natl Acad Sci U S A ; 113(29): 8314-9, 2016 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-27364007

RESUMO

Inositol pyrophosphates such as 5-diphosphoinositol pentakisphosphate (5-IP7) are highly energetic inositol metabolites containing phosphoanhydride bonds. Although inositol pyrophosphates are known to regulate various biological events, including growth, survival, and metabolism, the molecular sites of 5-IP7 action in vesicle trafficking have remained largely elusive. We report here that elevated 5-IP7 levels, caused by overexpression of inositol hexakisphosphate (IP6) kinase 1 (IP6K1), suppressed depolarization-induced neurotransmitter release from PC12 cells. Conversely, IP6K1 depletion decreased intracellular 5-IP7 concentrations, leading to increased neurotransmitter release. Consistently, knockdown of IP6K1 in cultured hippocampal neurons augmented action potential-driven synaptic vesicle exocytosis at synapses. Using a FRET-based in vitro vesicle fusion assay, we found that 5-IP7, but not 1-IP7, exhibited significantly higher inhibitory activity toward synaptic vesicle exocytosis than IP6 Synaptotagmin 1 (Syt1), a Ca(2+) sensor essential for synaptic membrane fusion, was identified as a molecular target of 5-IP7 Notably, 5-IP7 showed a 45-fold higher binding affinity for Syt1 compared with IP6 In addition, 5-IP7-dependent inhibition of synaptic vesicle fusion was abolished by increasing Ca(2+) levels. Thus, 5-IP7 appears to act through Syt1 binding to interfere with the fusogenic activity of Ca(2+) These findings reveal a role of 5-IP7 as a potent inhibitor of Syt1 in controlling the synaptic exocytotic pathway and expand our understanding of the signaling mechanisms of inositol pyrophosphates.


Assuntos
Exocitose/efeitos dos fármacos , Fosfatos de Inositol/farmacologia , Sinaptotagmina I/fisiologia , Animais , Hipocampo/citologia , Neurônios/fisiologia , Células PC12 , Fosfotransferases (Aceptor do Grupo Fosfato)/metabolismo , Ratos , Ratos Sprague-Dawley
7.
J Biol Chem ; 290(16): 10535-43, 2015 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-25716321

RESUMO

In neuroexocytosis, SNAREs and Munc18-1 may consist of the minimal membrane fusion machinery. Consistent with this notion, we observed, using single molecule fluorescence assays, that Munc18-1 stimulates SNARE zippering and SNARE-dependent lipid mixing in the absence of a major Ca(2+) sensor synaptotagmin-1 (Syt1), providing the structural basis for the conserved function of Sec1/Munc18 proteins in exocytosis. However, when full-length Syt1 is present, no enhancement of SNARE zippering and no acceleration of Ca(2+)-triggered content mixing by Munc18-1 are observed. Thus, our results show that Syt1 acts as an antagonist for Munc18-1 in SNARE zippering and fusion pore opening. Although the Sec1/Munc18 family may serve as part of the fusion machinery in other exocytotic pathways, Munc18-1 may have evolved to play a different role, such as regulating syntaxin-1a in neuroexocytosis.


Assuntos
Cálcio/metabolismo , Proteínas Munc18/genética , Vesículas Sinápticas/metabolismo , Sinaptotagmina I/genética , Animais , Ligação Competitiva , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Cinética , Fusão de Membrana , Proteínas Munc18/metabolismo , Ligação Proteica , Proteolipídeos/química , Proteolipídeos/metabolismo , Ratos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transmissão Sináptica , Vesículas Sinápticas/química , Proteína 25 Associada a Sinaptossoma/genética , Proteína 25 Associada a Sinaptossoma/metabolismo , Sinaptotagmina I/metabolismo , Sintaxina 1/genética , Sintaxina 1/metabolismo , Proteína 2 Associada à Membrana da Vesícula/genética , Proteína 2 Associada à Membrana da Vesícula/metabolismo
8.
J Am Chem Soc ; 138(13): 4512-21, 2016 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-26987363

RESUMO

Membrane fusion is mediated by the SNARE complex which is formed through a zippering process. Here, we developed a chemical controller for the progress of membrane fusion. A hemifusion state was arrested by a polyphenol myricetin which binds to the SNARE complex. The arrest of membrane fusion was rescued by an enzyme laccase that removes myricetin from the SNARE complex. The rescued hemifusion state was metastable and long-lived with a decay constant of 39 min. This membrane fusion controller was applied to delineate how Ca(2+) stimulates fusion-pore formation in a millisecond time scale. We found, using a single-vesicle fusion assay, that such myricetin-primed vesicles with synaptotagmin 1 respond synchronously to physiological concentrations of Ca(2+). When 10 µM Ca(2+) was added to the hemifused vesicles, the majority of vesicles rapidly advanced to fusion pores with a time constant of 16.2 ms. Thus, the results demonstrate that a minimal exocytotic membrane fusion machinery composed of SNAREs and synaptotagmin 1 is capable of driving membrane fusion in a millisecond time scale when a proper vesicle priming is established. The chemical controller of SNARE-driven membrane fusion should serve as a versatile tool for investigating the differential roles of various synaptic proteins in discrete fusion steps.


Assuntos
Cálcio/metabolismo , Proteínas SNARE/metabolismo , Animais , Exocitose , Flavonoides/metabolismo , Lacase/metabolismo , Fusão de Membrana , Proteínas do Tecido Nervoso/metabolismo , Ligação Proteica , Ratos , Sinaptotagmina I/metabolismo
9.
Proc Natl Acad Sci U S A ; 110(10): 4087-92, 2013 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-23431141

RESUMO

Parkinson disease and dementia with Lewy bodies are featured with the formation of Lewy bodies composed mostly of α-synuclein (α-Syn) in the brain. Although evidence indicates that the large oligomeric or protofibril forms of α-Syn are neurotoxic agents, the detailed mechanisms of the toxic functions of the oligomers remain unclear. Here, we show that large α-Syn oligomers efficiently inhibit neuronal SNARE-mediated vesicle lipid mixing. Large α-Syn oligomers preferentially bind to the N-terminal domain of a vesicular SNARE protein, synaptobrevin-2, which blocks SNARE-mediated lipid mixing by preventing SNARE complex formation. In sharp contrast, the α-Syn monomer has a negligible effect on lipid mixing even with a 30-fold excess compared with the case of large α-Syn oligomers. Thus, the results suggest that large α-Syn oligomers function as inhibitors of dopamine release, which thus provides a clue, at the molecular level, to their neurotoxicity.


Assuntos
Neurônios/fisiologia , Proteínas SNARE/fisiologia , alfa-Sinucleína/química , alfa-Sinucleína/fisiologia , Animais , Exocitose/efeitos dos fármacos , Exocitose/fisiologia , Metabolismo dos Lipídeos/efeitos dos fármacos , Modelos Neurológicos , Neurônios/efeitos dos fármacos , Neurotoxinas/química , Neurotoxinas/toxicidade , Células PC12 , Ligação Proteica , Estrutura Quaternária de Proteína , Proteolipídeos/metabolismo , Ratos , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/toxicidade , Vesículas Secretórias/efeitos dos fármacos , Vesículas Secretórias/fisiologia , Transdução Genética , Proteína 2 Associada à Membrana da Vesícula/fisiologia , alfa-Sinucleína/genética , alfa-Sinucleína/toxicidade
10.
Biochem Biophys Res Commun ; 465(4): 864-70, 2015 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-26319432

RESUMO

Soluble N-ethylmaleimide-sensitive-factor attachment protein receptor (SNARE) proteins generate energy required for membrane fusion. They form a parallelly aligned four-helix bundle called the SNARE complex, whose formation is initiated from the N terminus and proceeds toward the membrane-proximal C terminus. Previously, we have shown that this zippering-like process can be controlled by several flavonoids that bind to the intermediate structures formed during the SNARE zippering. Here, our aim was to test whether the fluorescence resonance energy transfer signals that are observed during the inner leaflet mixing assay indeed represent the hemifused vesicles. We show that changes in vesicle size accompanying the merging of bilayers is a good measure of progression of the membrane fusion. Two merging vesicles with the same size D in diameter exhibited their hydrodynamic diameters 2D + d (d, intermembrane distance), 2D and 2D as membrane fusion progressed from vesicle docking to hemifusion and full fusion, respectively. A dynamic light scattering assay of membrane fusion suggested that myricetin stopped membrane fusion at the hemifusion state, whereas delphinidin and cyanidin prevented the docking of the vesicles. These results are consistent with our previous findings in fluorescence resonance energy transfer assays.


Assuntos
Flavonoides/metabolismo , Fusão de Membrana/fisiologia , Proteínas SNARE/química , Proteínas SNARE/metabolismo , Animais , Difusão Dinâmica da Luz , Flavonoides/farmacologia , Transferência Ressonante de Energia de Fluorescência , Hidrodinâmica , Bicamadas Lipídicas/metabolismo , Fusão de Membrana/efeitos dos fármacos , Complexos Multiproteicos/química , Complexos Multiproteicos/metabolismo , Ligação Proteica , Ratos , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
11.
Biophys J ; 107(7): 1601-8, 2014 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-25296312

RESUMO

In Alzheimer's disease, cytochrome c-dependent apoptosis is a crucial pathway in neuronal cell death. Although beta-amyloid (Aß) oligomers are known to be the neurotoxins responsible for neuronal cell death, the underlying mechanisms remain largely elusive. Here, we report that the oligomeric form of synthetic Aß of 42 amino acids elicits death of HT-22 cells. But, when expression of a bcl-2 family protein BAK is suppressed by siRNA, Aß oligomer-induced cell death was reduced. Furthermore, significant reduction of cytochrome c release was observed with mitochondria isolated from BAK siRNA-treated HT-22 cells. Our in vitro experiments demonstrate that Aß oligomers bind to BAK on the membrane and induce apoptotic BAK pores and cytochrome c release. Thus, the results suggest that Aß oligomers function as apoptotic ligands and hijack the intrinsic apoptotic pathway to cause unintended neuronal cell death.


Assuntos
Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/farmacologia , Apoptose/efeitos dos fármacos , Citocromos c/metabolismo , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/farmacologia , Multimerização Proteica , Proteína Killer-Antagonista Homóloga a bcl-2/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/metabolismo , Linhagem Celular , Camundongos , Fragmentos de Peptídeos/metabolismo , Porosidade , Estrutura Secundária de Proteína , alfa-Sinucleína/farmacologia , Proteínas tau/farmacologia
12.
Biochem Biophys Res Commun ; 450(1): 831-6, 2014 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-24960195

RESUMO

Fusion of synaptic vesicles with the presynaptic plasma membrane in the neuron is mediated by soluble N-ethylmaleimide-sensitive fusion protein-attachment protein receptor (SNARE) proteins. SNARE complex formation is a zippering-like process which initiates at the N-terminus and proceeds to the C-terminal membrane-proximal region. Previously, we showed that this zippering-like process is regulated by several polyphenols, leading to the arrest of membrane fusion and the inhibition of neuroexocytosis. In vitro studies using purified SNARE proteins reconstituted in liposomes revealed that each polyphenol uniquely regulates SNARE zippering. However, the unique regulatory effect of each polyphenol in cells has not yet been examined. In the present study, we observed SNARE zippering in neuronal PC12 cells by measuring the fluorescence resonance energy transfer (FRET) changes of a cyan fluorescence protein (CFP) and a yellow fluorescence protein (YFP) fused to the N-termini or C-termini of SNARE proteins. We show that delphinidin and cyanidin inhibit the initial N-terminal nucleation of SNARE complex formation in a Ca(2+)-independent manner, while myricetin inhibits Ca(2+)-dependent transmembrane domain association of the SNARE complex in the cell. This result explains how polyphenols exhibit botulinum neurotoxin-like activity in vivo.


Assuntos
Cálcio/metabolismo , Membrana Celular/metabolismo , Zíper de Leucina/efeitos dos fármacos , Fusão de Membrana/fisiologia , Neurônios/metabolismo , Polifenóis/farmacologia , Proteínas SNARE/metabolismo , Animais , Membrana Celular/efeitos dos fármacos , Fusão de Membrana/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Células PC12 , Ratos
13.
Biochem J ; 450(3): 537-46, 2013 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-23252429

RESUMO

Anti-allergic effects of dietary polyphenols were extensively studied in numerous allergic disease models, but the molecular mechanisms of anti-allergic effects by polyphenols remain poorly understood. In the present study, we show that the release of granular cargo molecules, contained in distinct subsets of granules of mast cells, is specifically mediated by two sets of SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptor) proteins, and that various polyphenols differentially inhibit the formation of those SNARE complexes. Expression analysis of RBL-2H3 cells for 11 SNARE genes and a lipid mixing assay of 24 possible combinations of reconstituted SNAREs indicated that the only two active SNARE complexes involved in mast cell degranulation are Syn (syntaxin) 4/SNAP (23 kDa synaptosome-associated protein)-23/VAMP (vesicle-associated membrane protein) 2 and Syn4/SNAP-23/VAMP8. Various polyphenols selectively or commonly interfered with ternary complex formation of these two SNARE complexes, thereby stopping membrane fusion between granules and plasma membrane. This led to the differential effect of polyphenols on degranulation of three distinct subsets of granules. These results suggest the possibility that formation of a variety of SNARE complexes in numerous cell types is controlled by polyphenols which, in turn, might regulate corresponding membrane trafficking.


Assuntos
Degranulação Celular/efeitos dos fármacos , Mastócitos/efeitos dos fármacos , Polifenóis/farmacologia , Proteínas SNARE/metabolismo , Vesículas Transportadoras/efeitos dos fármacos , Células Cultivadas , Grânulos Citoplasmáticos/metabolismo , Regulação para Baixo/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Histamina/metabolismo , Humanos , Mastócitos/metabolismo , Mastócitos/fisiologia , Complexos Multiproteicos/metabolismo , Polifenóis/metabolismo , Ligação Proteica/efeitos dos fármacos , Especificidade por Substrato/efeitos dos fármacos , Vesículas Transportadoras/classificação , Vesículas Transportadoras/fisiologia , beta-N-Acetil-Hexosaminidases/metabolismo
14.
Bioact Mater ; 34: 138-149, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38223538

RESUMO

Oral administration facilitates the direct delivery of drugs to lesions within the small intestine and colon, making it an ideal approach for treating patients with inflammatory bowel disease. However, multiple physical barriers impede the delivery of oral RNA drugs through the gastrointestinal tract. Herein, we developed a novel oral siRNA delivery system that protects nucleic acids in extreme environments by employing exosomes derived from milk to encapsulate tumor necrosis factor-alpha (TNF-α) siRNA completely. The remarkable structural stability of milk-derived exosomes (M-Exos), as opposed to those from HEK293T cells, makes them exceptional siRNA carriers. Results demonstrate that milk exosomes loaded with TNF-α siRNA (M-Exo/siR) can effectively inhibit the expression of TNF-α-related inflammatory cytokines. Moreover, given that milk exosomes are composed of unique lipids with high bioavailability, orally administered M-Exo/siR effectively reach colonic tissues, leading to decreased TNF-α expression and successful alleviation of colitis symptoms in a dextran sulfate sodium-induced inflammatory bowel disease murine model. Hence, milk-derived exosomes carrying TNF-α siRNA can be effectively employed to treat inflammatory bowel disease. Indeed, using exosomes naturally derived from milk may shift the current paradigm of oral gene delivery, including siRNA.

15.
Adv Mater ; : e2405475, 2024 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-38898702

RESUMO

While proteolysis-targeting chimeras (PROTACs) hold great potential for persistently reprogramming the immunosuppressive tumor microenvironment via targeted protein degradation, precisely activating them in tumor tissues and preventing uncontrolled proteolysis at off-target sites remain challenging. Herein, a light-triggered PROTAC nanoassembly (LPN) for photodynamic indoleamine 2,3-dioxygenase (IDO) proteolysis is reported. The LPN is derived from the self-assembly of prodrug conjugates, which comprise a PROTAC, cathepsin B-specific cleavable peptide linker, and photosensitizer, without any additional carrier materials. In colon tumor models, intravenously injected LPNs initially silence the activity of PROTACs and accumulate significantly in targeted tumor tissues due to an enhanced permeability and retention effect. Subsequently, the cancer biomarker cathepsin B begins to trigger the release of active PROTACs from the LPNs through enzymatic cleavage of the linkers. Upon light irradiation, tumor cells undergo immunogenic cell death induced by photodynamic therapy to promote the activation of effector T cells, while the continuous IDO degradation of PROTAC simultaneously blocks tryptophan metabolite-regulated regulatory-T-cell-mediated immunosuppression. Such LPN-mediated combinatorial photodynamic IDO proteolysis effectively inhibits tumor growth, metastasis, and recurrence. Collectively, this study presents a promising nanomedicine, designed to synergize PROTACs with other immunotherapeutic modalities, for more effective and safer cancer immunotherapy.

16.
ACS Nano ; 18(25): 16297-16311, 2024 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-38867457

RESUMO

While mesalamine, a 5-aminosalicylic acid (5-ASA), is pivotal in the management of inflammatory bowel disease (IBD) through both step-up and top-down approaches in clinical settings, its widespread utilization is limited by low bioavailability at the desired site of action due to rapid and extensive absorption in the upper gastrointestinal (GI) tract. Addressing mesalamine's pharmacokinetic challenges, here, we introduce nanoassemblies composed exclusively of a mesalamine prodrug that pairs 5-ASA with a mucoadhesive and cathepsin B-cleavable peptide. In an IBD model, orally administered nanoassemblies demonstrate enhanced accumulation and sustained retention in the GI tract due to their mucoadhesive properties and the epithelial enhanced permeability and retention (eEPR) effect. This retention enables the efficient uptake by intestinal pro-inflammatory macrophages expressing high cathepsin B, triggering a burst release of the 5-ASA. This cascade fosters the polarization toward an M2 macrophage phenotype, diminishes inflammatory responses, and simultaneously facilitates the delivery of active agents to adjacent epithelial cells. Therefore, the nanoassemblies show outstanding therapeutic efficacy in inhibiting local inflammation and contribute to suppressing systemic inflammation by restoring damaged intestinal barriers. Collectively, this study highlights the promising role of the prodrug nanoassemblies in enhancing targeted drug delivery, potentially broadening the use of mesalamine in managing IBD.


Assuntos
Doenças Inflamatórias Intestinais , Macrófagos , Mesalamina , Pró-Fármacos , Mesalamina/química , Mesalamina/farmacologia , Pró-Fármacos/química , Pró-Fármacos/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Doenças Inflamatórias Intestinais/tratamento farmacológico , Animais , Camundongos , Humanos , Nanopartículas/química , Mucosa Intestinal/metabolismo , Mucosa Intestinal/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Anti-Inflamatórios não Esteroides/química , Anti-Inflamatórios não Esteroides/farmacologia , Anti-Inflamatórios não Esteroides/administração & dosagem
17.
Adv Sci (Weinh) ; 11(22): e2309917, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38520717

RESUMO

Lipid nanoparticles (LNPs) exhibit remarkable mRNA delivery efficiency, yet their majority accumulate in the liver or spleen after injection. Tissue-specific mRNA delivery can be achieved through modulating LNP properties, such as tuning PEGylation or varying lipid components systematically. In this paper, a streamlined method is used for incorporating tumor-targeting peptides into the LNPs; the programmed death ligand 1 (PD-L1) binding peptides are conjugated to PEGylated lipids via a copper-free click reaction, and directly incorporated into the LNP composition (Pep LNPs). Notably, Pep LNPs display robust interaction with PD-L1 proteins, which leads to the uptake of LNPs into PD-L1 overexpressing cancer cells both in vitro and in vivo. To evaluate anticancer immunotherapy mediated by restoring tumor suppressor, mRNA encoding phosphatase and tensin homolog (PTEN) is delivered via Pep LNPs to PTEN-deficient triple-negative breast cancers (TNBCs). Pep LNPs loaded with PTEN mRNA specifically promotes autophagy-mediated immunogenic cell death in 4T1 tumors, resulting in effective anticancer immune responses. This study highlights the potential of tumor-targeted LNPs for mRNA-based cancer therapy.


Assuntos
Antígeno B7-H1 , Nanopartículas , PTEN Fosfo-Hidrolase , PTEN Fosfo-Hidrolase/metabolismo , PTEN Fosfo-Hidrolase/genética , Nanopartículas/química , Animais , Camundongos , Antígeno B7-H1/metabolismo , Antígeno B7-H1/genética , Feminino , Modelos Animais de Doenças , Lipídeos/química , Humanos , Linhagem Celular Tumoral , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/terapia , Camundongos Endogâmicos BALB C , Imunoterapia/métodos , Lipossomos
18.
Proc Natl Acad Sci U S A ; 107(51): 22145-50, 2010 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-21135223

RESUMO

Neuronal SNARE proteins mediate neurotransmitter release at the synapse by facilitating the fusion of vesicles to the presynaptic plasma membrane. Cognate v-SNAREs and t-SNAREs from the vesicle and the plasma membrane, respectively, zip up and bring about the apposition of two membranes attached at the C-terminal ends. Here, we demonstrate that SNARE zippering can be modulated in the midways by wedging with small hydrophobic molecules. Myricetin, which intercalated into the hydrophobic inner core near the middle of the SNARE complex, stopped SNARE zippering in motion and accumulated the trans-complex, where the N-terminal region of v-SNARE VAMP2 is in the coiled coil with the frayed C-terminal region. Delphinidin and cyanidin inhibited N-terminal nucleation of SNARE zippering. Neuronal SNARE complex in PC12 cells showed the same pattern of vulnerability to small hydrophobic molecules. We propose that the half-zipped trans-SNARE complex is a crucial intermediate waiting for a calcium trigger that leads to fusion pore opening.


Assuntos
Cálcio/metabolismo , Membrana Celular/metabolismo , Fusão de Membrana/fisiologia , Proteínas SNARE/metabolismo , Animais , Antocianinas/farmacologia , Membrana Celular/genética , Flavonoides/farmacologia , Interações Hidrofóbicas e Hidrofílicas/efeitos dos fármacos , Fusão de Membrana/efeitos dos fármacos , Células PC12 , Estrutura Terciária de Proteína , Ratos , Proteínas SNARE/genética
19.
Adv Drug Deliv Rev ; 199: 114993, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37414361

RESUMO

Messenger RNA (mRNA) is now in the limelight as a powerful tool for treating various human diseases, especially malignant tumors, thanks to the remarkable clinical outcomes of mRNA vaccines using lipid nanoparticle technology during the COVID-19 pandemic. Recent promising preclinical and clinical results that epitomize the advancement in mRNA and nanoformulation-based delivery technologies have highlighted the tremendous potential of mRNA in cancer immunotherapy. mRNAs can be harnessed for cancer immunotherapy in forms of various therapeutic modalities, including cancer vaccines, adoptive T-cell therapies, therapeutic antibodies, and immunomodulatory proteins. This review provides a comprehensive overview of the current state and prospects of mRNA-based therapeutics, including numerous delivery and therapeutic strategies.


Assuntos
COVID-19 , Neoplasias , Humanos , RNA Mensageiro , Pandemias , COVID-19/terapia , Imunoterapia/métodos
20.
J Control Release ; 359: 85-96, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37230294

RESUMO

Immune checkpoint blockade (ICB) therapy has shown remarkable outcomes along with multiple cases of complete regression in clinical practice. But unfortunately, most patients who have an immunosuppressive tumor immune microenvironment (TIME) respond poorly to these therapies. To improve the response rate of the patients, various treatment modalities that can boost cancer immunogenicity and remove immune tolerance have been combined with ICB therapies. However, the systemic administration of multiple immunotherapeutic agents can potentially cause severe off-target toxicities and immune-related adverse events, diminishing antitumor immunity and increasing the risk of additional complications. To address these problems, Immune Checkpoint-Targeted Drug Conjugates (IDCs) have been widely investigated for their ability to offer distinct advantages in remodeling the TIME for cancer immunotherapy. IDCs, consisting of immune checkpoint-targeting moieties, cleavable linkers, and payloads of immunotherapeutic agents, have a similar structure to conventional antibody-drug conjugates (ADCs) but target and block the immune checkpoint receptors, and then release the payloads conjugated through cleavable linkers. These unique mechanisms of IDCs prompt an immune-responsive TIME by modulating the multiple steps related to the cancer-immunity cycle, ultimately leading to tumor eradication. This review outlines the mode of action and advantages of IDCs. In addition, various IDCs for combinational immunotherapy are reviewed. Finally, the potential and challenges of IDCs for clinical translation are discussed.


Assuntos
Neoplasias , Humanos , Neoplasias/tratamento farmacológico , Fatores Imunológicos , Imunoterapia , Microambiente Tumoral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA