Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Molecules ; 26(1)2020 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-33375296

RESUMO

The advance of structural biology has revealed numerous noncovalent interactions between peptide sequences in protein structures, but such information is less explored for developing peptide materials. Here we report the formation of heterotypic peptide hydrogels by the two binding motifs revealed by the structures of an inflammasome. Specifically, conjugating a self-assembling motif to the positively or negatively charged peptide sequence from the ASCPYD filaments of inflammasome produces the solutions of the peptides. The addition of the peptides of the oppositely charged and complementary peptides to the corresponding peptide solution produces the heterotypic hydrogels. Rheology measurement shows that ratios of the complementary peptides affect the viscoelasticity of the resulted hydrogel. Circular dichroism indicates that the addition of the complementary peptides results in electrostatic interactions that modulate self-assembly. Transmission electron microscopy reveals that the ratio of the complementary peptides controls the morphology of the heterotypic peptide assemblies. This work illustrates a rational, biomimetic approach that uses the structural information from the protein data base (PDB) for developing heterotypic peptide materials via self-assembly.


Assuntos
Hidrogéis/química , Inflamassomos/metabolismo , Dicroísmo Circular , Módulo de Elasticidade , Inflamassomos/ultraestrutura , Modelos Moleculares , Imagem Óptica , Transição de Fase
2.
J Am Chem Soc ; 141(18): 7271-7274, 2019 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-31033285

RESUMO

Cell-mediated remodeling of extracellular matrix (ECM) plays important roles for cell functions, but it is challenging to develop synthetic materials for mimicking such a dynamic aspect of proteins in ECM. Here we show that intercellular morphological transition of peptide assemblies mimic the unfolding of fibronectin, thus enabling formation of spheroids from a monolayer of cells. Specifically, the phosphopeptide self-assembles to form nanoparticles, which turns into nanofibers upon partial dephosphorylation catalyzed by enzymes (e.g., phosphatases) at intercellular space. Occurring between HS-5 cells, such an enzyme-instructed self-assembly enables a sheet of the HS-5 cells to form cell spheroids. Structure-activity investigation reveals that proteolytic stability, dephosphorylation, and biotin conjugation of the peptides are indispensable for forming the cell spheroids. Further mechanism study indicates that the intercellular assemblies interact with multiple ECM components (e.g., laminin, collagens III and IV) to drive the formation of the cell spheroids. As the first example of intercellular instructed-assembly from homotypic precursors, this work illustrates a new approach that uses cell-responsive peptide assemblies to mimic protein dynamics for control cell behaviors.


Assuntos
Proteínas da Matriz Extracelular/metabolismo , Esferoides Celulares/metabolismo , Linhagem Celular , Matriz Extracelular/química , Matriz Extracelular/metabolismo , Proteínas da Matriz Extracelular/química , Humanos , Conformação Molecular , Tamanho da Partícula , Esferoides Celulares/química , Propriedades de Superfície
3.
Chembiochem ; 20(19): 2442-2446, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30957316

RESUMO

Biological systems have evolved to create a structural and dynamic continuum of bio-macromolecular assemblies for the purpose of optimizing the system's functions. The formation of these dynamic higher-order assemblies is precisely controlled by biological cues. However, controlling the self-assembly of synthetic molecules spatiotemporally in or on live cells is still a big challenge, especially for performing functions. This concept article introduces the use of in situ reactions as a spatiotemporal control to form assemblies of small molecules that induce cell morphogenesis or apoptosis. After briefly introducing a representative example of a natural dynamic continuum of the higher-order assemblies, we describe enzyme-instructed self-assembly (EISA) for constructing dynamic assemblies of small molecules, then discuss the use of EISA for controlling cell morphogenesis and apoptosis. Finally, we provide a brief outlook to discuss the future perspective of this exciting new research direction.


Assuntos
Células da Medula Óssea/citologia , Neoplasias Ósseas/patologia , Substâncias Macromoleculares/metabolismo , Osteossarcoma/patologia , Bibliotecas de Moléculas Pequenas/metabolismo , Células Estromais/citologia , Células da Medula Óssea/metabolismo , Neoplasias Ósseas/metabolismo , Linhagem da Célula , Células Cultivadas , Humanos , Substâncias Macromoleculares/química , Osteossarcoma/metabolismo , Bibliotecas de Moléculas Pequenas/química , Esferoides Celulares/metabolismo , Células Estromais/metabolismo
4.
Bioconjug Chem ; 30(10): 2528-2532, 2019 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-31550136

RESUMO

Selectively targeting the cell nucleolus remains a challenge. Here, we report the first case in which d-peptides form membraneless molecular condensates with RNA for targeting cell nucleolus. A d-peptide derivative, enriched with lysine and hydrophobic residues, self-assembles to form nanoparticles, which enter cells through clathrin-dependent endocytosis and mainly accumulate at the cell nucleolus. A structural analogue of the d-peptide reveals that the particle morphology of the assemblies, which depends on the side chain modification, favors the cellular uptake. In contrast to those of the d-peptide, the assemblies of the corresponding l-enantiomer largely localize in cell lysosomes. Preliminary mechanism study suggests that the d-peptide nanoparticles interact with RNA to form membraneless condensates in the nucleolus, which further induces DNA damage and results in cell death. This work illustrates a new strategy for rationally designing supramolecular assemblies of d-peptides for targeting subcellular organelles.


Assuntos
Nucléolo Celular/metabolismo , Peptídeos/metabolismo , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Humanos , Imagem Óptica , Peptídeos/química
5.
Angew Chem Int Ed Engl ; 58(31): 10423-10432, 2019 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-30903643

RESUMO

Using peptide assemblies with emergent properties to achieve elaborate functions has attracted increasing attention in recent years. Besides tailoring the self-assembly of peptides in vitro, peptide research is advancing into a new and exciting frontier: the rational design of peptide assemblies (or their derivatives) for biological functions in a complex environment. This Minireview highlights recent developments in peptide assemblies and their applications in biological systems. After introducing the unique merits of peptide assemblies, we discuss the recent progress in designing peptides (or peptide derivatives) for self-assembly with conformational control. Then, we describe biological functions of peptide assemblies, with an emphasis on approach-instructed assembly for spatiotemporal control of peptide assemblies, in the cellular context. Finally, we discuss the future promises and challenges of this exciting area of chemistry.


Assuntos
Peptídeos/síntese química , Humanos , Estrutura Molecular , Tamanho da Partícula , Peptídeos/química , Peptídeos/metabolismo , Conformação Proteica , Propriedades de Superfície
6.
Angew Chem Int Ed Engl ; 58(17): 5567-5571, 2019 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-30801914

RESUMO

Context-dependent signaling is a ubiquitous phenomenon in nature, but ways to mimic the essence of these nano- and microscale dynamic molecular processes by noncovalent synthesis in the cellular environment have yet to be developed. Herein we present a dynamic continuum of noncovalent filaments formed by the instructed assembly (iA) of a supramolecular phosphoglycopeptide (sPGP) as context-dependent signals for controlling the death and morphogenesis of cells. Specifically, ectophosphatase enzymes on cancer cells catalyze the formation of sPGP filaments to result in cell death; however, damping of the enzyme activity induces the formation 3D cell spheroids. Similarly, the ratio of stromal and cancer cells in a coculture can be used to modulate the expression of the ectophosphatase, so that the iA process leads to the formation of cell spheroids. The spheroids mimic the tumor microenvironment for drug screening.


Assuntos
Movimento Celular , Humanos , Transdução de Sinais
7.
J Am Chem Soc ; 140(48): 16433-16437, 2018 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-30452246

RESUMO

Liquid-like droplets of biomacromolecules are emerging as a fundamental mechanism of cellular signaling, but designing synthetic mimics to form such membraneless organelles remains unexplored. Here we report the use of supramolecular assemblies of small peptides, as a mimic of biomacromolecular condensates, for intracellular sequestration of enzymes on endoplasmic reticulum (ER). Specifically, integrating a short peptide with naproxen (a nonsteroidal anti-inflammatory drug (NSAID) and a ligand of cyclooxygenase-2 (COX-2)) generates an enzymatic substrate that acts as a precursor for instructed assembly. Slowly dephosphorylating the precursors by phosphatases forms the corresponding hydrogelators in a cellular environment, which results in the supramolecular assemblies on ER. Consisting of the precursor and the hydrogelator molecules, the assemblies enable the sequestration of COX-2 and protein-tyrosine phosphatase 1B (PTP1B) on ER. Further structure-activity investigation reveals that the colocalization of COX-2 and PTP1B relies on the NSAID motif, the phosphotyrosine, and the enzymatic dephosphorylation of the precursor. This work, for the first time, illustrates the use of supramolecular processes for associating enzymes in cells and may provide insights for understanding intracellular liquid condensates and a new strategy for modulating protein-protein interactions.


Assuntos
Anti-Inflamatórios não Esteroides/metabolismo , Inibidores de Ciclo-Oxigenase 2/metabolismo , Ciclo-Oxigenase 2/metabolismo , Naproxeno/metabolismo , Oligopeptídeos/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 1/metabolismo , Anti-Inflamatórios não Esteroides/química , Linhagem Celular Tumoral , Inibidores de Ciclo-Oxigenase 2/química , Retículo Endoplasmático/metabolismo , Humanos , Hidrogéis/química , Hidrogéis/metabolismo , Naproxeno/química , Oligopeptídeos/síntese química , Oligopeptídeos/química
8.
J Am Chem Soc ; 140(10): 3505-3509, 2018 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-29481071

RESUMO

Despite the advancement of molecular imaging techniques, there is an unmet need for probes for direct imaging of membrane dynamics of live cells. Here we report a novel type of active (or enzyme responsive) probes to directly image membrane dynamics of live cells with high spatial and temporal resolution over extended time scales and areas. Because lipid rafts enrich cholesterols and GPI-anchored enzymes (e.g., ectophosphatases), we design probes that consist of an enzymatic trigger, a fluorophore, and a cholesterol that are affinitive to the cell membrane. Being water-soluble and as the substrate of ectophosphatase, these cell compatible probes preferentially and rapidly assemble in plasma membrane, exhibit strong fluorescence, work at micromolar concentrations, and easily achieve high resolution monitoring of nanoscale heterogeneity in membranes of live cells, the release of exosomes, and the membrane dynamics of live cells. This work provides a facile means to link membrane dynamics and heterogeneity to cellular processes for understanding the interactions between membranes and proteins.


Assuntos
Membrana Celular/metabolismo , Corantes Fluorescentes/química , Imagem Molecular , Linhagem Celular , Membrana Celular/química , Sobrevivência Celular , Humanos , Estrutura Molecular
9.
J Am Chem Soc ; 140(30): 9566-9573, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29995402

RESUMO

The endoplasmic reticulum (ER) is responsible for the synthesis and folding of a large number of proteins, as well as intracellular calcium regulation, lipid synthesis, and lipid transfer to other organelles, and is emerging as a target for cancer therapy. However, strategies for selectively targeting the ER of cancer cells are limited. Here we show that enzymatically generated crescent-shaped supramolecular assemblies of short peptides disrupt cell membranes and target ER for selective cancer cell death. As revealed by sedimentation assay, the assemblies interact with synthetic lipid membranes. Live cell imaging confirms that the assemblies impair membrane integrity, which is further supported by lactate dehydrogenase (LDH) assays. According to transmission electron microscopy (TEM), static light scattering (SLS), and critical micelle concentration (CMC), attaching an l-amino acid at the C-terminal of a d-tripeptide results in the crescent-shaped supramolecular assemblies. Structure-activity relationship suggests that the crescent-shaped morphology is critical for interacting with membranes and for controlling cell fate. Moreover, fluorescent imaging indicates that the assemblies accumulate on the ER. Time-dependent Western blot and ELISA indicate that the accumulation causes ER stress and subsequently activates the caspase signaling cascade for cell death. As an approach for in situ generating membrane binding scaffolds (i.e., the crescent-shaped supramolecular assemblies), this work promises a new way to disrupt the membrane and to target the ER for developing anticancer therapeutics.


Assuntos
Antineoplásicos/farmacologia , Membrana Celular/metabolismo , Retículo Endoplasmático/metabolismo , Oligopeptídeos/farmacologia , Fosfopeptídeos/farmacologia , Fosfatase Alcalina/metabolismo , Antineoplásicos/síntese química , Antineoplásicos/química , Antineoplásicos/metabolismo , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Humanos , Lipossomos/metabolismo , Estrutura Molecular , Oligopeptídeos/síntese química , Oligopeptídeos/química , Oligopeptídeos/metabolismo , Fosfopeptídeos/síntese química , Fosfopeptídeos/química , Fosfopeptídeos/metabolismo , Multimerização Proteica , Relação Estrutura-Atividade
10.
Chem Soc Rev ; 46(9): 2421-2436, 2017 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-28357433

RESUMO

Self-assembly, the autonomous organization of components to form patterns or structures, is a prevalent process in nature at all scales. Particularly, biological systems offer remarkable examples of diverse structures (as well as building blocks) and processes resulting from self-assembly. The exploration of bioinspired assemblies not only allows for mimicking the structures of living systems, but it also leads to functions for applications in different fields that benefit humans. In the last several decades, efforts on understanding and controlling self-assembly of small molecules have produced a large library of candidates for developing the biomedical applications of assemblies of small molecules. Moreover, recent findings in biology have provided new insights on the assemblies of small molecules to modulate essential cellular processes (such as apoptosis). These observations indicate that the self-assembly of small molecules, as multifaceted entities and processes to interact with multiple proteins, can have profound biological impacts on cells. In this review, we illustrate that the generation of assemblies of small molecules in cell milieu with their interactions with multiple cellular proteins for regulating cellular processes can result in primary phenotypes, thus providing a fundamentally new molecular approach for controlling cell behavior. By discussing the correlation between molecular assemblies in nature and the assemblies of small molecules in cell milieu, illustrating the functions of the assemblies of small molecules, and summarizing some guiding principles, we hope this review will stimulate more molecular scientists to explore the bioinspired self-assembly of small molecules in cell milieu.


Assuntos
Células/química , Células/metabolismo , Bibliotecas de Moléculas Pequenas/metabolismo , Humanos , Bibliotecas de Moléculas Pequenas/química
11.
Chem Soc Rev ; 46(21): 6470-6479, 2017 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-28849819

RESUMO

In this review, supramolecular catalysis refers to the integration of the catalytic process with molecular self-assembly driven by noncovalent interactions, and dynamic assemblies are the assemblies that form and dissipate reversibly. Cells extensively employ supramolecular catalysis and dynamic assemblies for controlling their complex functions. The dynamic generation of supramolecular assemblies of small molecules has made considerable progress in the last decade, though the disassembly processes remain underexplored. Here, we discuss the regulation of dynamic assemblies via self-assembly and disassembly processes for therapeutics and diagnostics. We first briefly introduce the self-assembly and disassembly processes in the context of cells, which provide the rationale for designing approaches to control the assemblies. Then, we describe recent advances in designing and regulating the self-assembly and disassembly of small molecules, especially for molecular imaging and anticancer therapeutics. Finally, we provide a perspective on future directions of the research on supramolecular catalysis and dynamic assemblies for medicine.


Assuntos
Antineoplásicos/química , Simulação de Dinâmica Molecular , Imagem Molecular , Neoplasias/tratamento farmacológico , Bibliotecas de Moléculas Pequenas/química , Antineoplásicos/síntese química , Antineoplásicos/farmacologia , Catálise , Proliferação de Células/efeitos dos fármacos , Humanos , Substâncias Macromoleculares/síntese química , Substâncias Macromoleculares/química , Substâncias Macromoleculares/farmacologia , Neoplasias/patologia , Bibliotecas de Moléculas Pequenas/síntese química , Bibliotecas de Moléculas Pequenas/farmacologia
12.
Angew Chem Int Ed Engl ; 57(18): 4931-4935, 2018 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-29451962

RESUMO

Herein, we report that assemblies of nucleopeptides selectively sequester ATP in complex conditions (for example, serum and cytosol). We developed assemblies of nucleopeptides that selectively sequester ATP over ADP. Counteracting enzymes interconvert ATP and ADP to modulate the nanostructures formed by the nucleopeptides and the nucleotides. The nucleopeptides, sequestering ATP effectively in cells, slow down efflux pumps in multidrug-resistant cancer cells, thus boosting the efficacy of doxorubicin, an anticancer drug. Investigation of 11 nucleopeptides (including d- and l-enantiomers) yields five more nucleopeptides that differentiate ATP and ADP through either precipitation or gelation. As the first example of assemblies of nucleopeptides that interact with ATP and disrupt intracellular ATP dynamics, this work illustrates the use of supramolecular assemblies to interact with small and essential biological molecules for controlling cell behavior.


Assuntos
Trifosfato de Adenosina/química , Antineoplásicos/farmacologia , Doxorrubicina/farmacologia , Peptídeos/síntese química , Antineoplásicos/química , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Doxorrubicina/química , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Modelos Moleculares , Estrutura Molecular , Peptídeos/química
13.
J Am Chem Soc ; 139(11): 3950-3953, 2017 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-28257192

RESUMO

Cancer cells differ from normal cells in both gain of functions (i.e., upregulation) and loss of functions (i.e., downregulation). While it is common to suppress gain of function for chemotherapy, it remains challenging to target downregulation in cancer cells. Here we show the combination of enzyme-instructed assembly and disassembly to target downregulation in cancer cells by designing peptidic precursors as the substrates of both carboxylesterases (CESs) and alkaline phosphatases (ALPs). The precursors turn into self-assembling molecules to form nanofibrils upon dephosphorylation by ALP, but CES-catalyzed cleavage of the ester bond on the molecules results in disassembly of the nanofibrils. The precursors selectively inhibit the cancer cells that downregulate CES (e.g., OVSAHO) but are innocuous to a hepatocyte that overexpresses CES (HepG2), while the two cell lines exhibit comparable ALP activities. This work illustrates a potential approach for the development of chemotherapy via targeting downregulation (or loss of functions) in cancer cells.


Assuntos
Fosfatase Alcalina/metabolismo , Hidrolases de Éster Carboxílico/metabolismo , Regulação para Baixo , Neoplasias Hepáticas/metabolismo , Neoplasias Ovarianas/metabolismo , Fosfatase Alcalina/química , Hidrolases de Éster Carboxílico/química , Linhagem Celular Tumoral , Feminino , Células Hep G2 , Humanos , Neoplasias Hepáticas/patologia , Neoplasias Ovarianas/patologia
14.
J Am Chem Soc ; 139(43): 15377-15384, 2017 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-28990765

RESUMO

Enzyme-instructed self-assembly (EISA) represents a dynamic continuum of supramolecular nanostructures that selectively inhibits cancer cells via simultaneously targeting multiple hallmark capabilities of cancer, but how to design the small molecules for EISA from the vast molecular space remains an unanswered question. Here we show that the self-assembling ability of small molecules controls the anticancer activity of EISA. Examining the EISA precursor analogues consisting of an N-capped d-tetrapeptide, a phosphotyrosine residue, and a diester or a diamide group, we find that, regardless of the stereochemistry and the regiochemistry of their tetrapeptidic backbones, the anticancer activities of these precursors largely match their self-assembling abilities. Additional mechanistic studies confirm that the assemblies of the small peptide derivatives result in cell death, accompanying significant rearrangement of cytoskeletal proteins and plasma membranes. These results imply that the diester or diamide derivatives of the d-tetrapeptides self-assemble pericellularly, as well as intracellularly, to result in cell death. As the first case to correlate thermodynamic properties (e.g., self-assembling ability) of small molecules with the efficacy of a molecule process against cancer cells, this work provides an important insight for developing a molecular dynamic continuum for potential cancer therapy, as well as understanding the cytotoxicity of pathogenic assemblies.


Assuntos
Morte Celular/efeitos dos fármacos , Nanoestruturas/química , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Peptídeos/síntese química , Peptídeos/uso terapêutico , Linhagem Celular Tumoral , Membrana Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Proteínas do Citoesqueleto/metabolismo , Humanos , Simulação de Dinâmica Molecular , Peptídeos/química , Peptídeos/farmacologia , Termodinâmica
15.
Angew Chem Int Ed Engl ; 56(26): 7579-7583, 2017 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-28481474

RESUMO

Based on the recent near-atomic structures of the PYRIN domain of ASC in the protein filament of inflammasomes and the observation that the active form of vitamin B6 (pyridoxal phosphate, P5P) modulates the self-assembly of ASC, we rationally designed an N-terminal capped nonapeptide (Nap-FFKKFKLKL, 1) to form supramolecular nanofibers consisting of α-helix. The addition of P5P to the solution of 1 results in a hydrogel almost instantly (about 4 seconds). Several other endogenous small molecules (for example, pyridoxal, folinic acid, ATP, and AMP) also convert the solution of 1 into a hydrogel. As the demonstration of correlating assemblies of peptides and the relevant protein epitopes, this work illustrates a bioinspired approach to develop supramolecular structures modulated by endogenous small molecules.


Assuntos
Hidrogéis/química , Inflamassomos/química , Sequência de Aminoácidos , Epitopos/química , Microscopia Eletrônica de Transmissão , Nanofibras/química , Oligopeptídeos/química , Conformação Proteica
16.
Angew Chem Int Ed Engl ; 56(51): 16297-16301, 2017 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-29125896

RESUMO

Higher-order assemblies of proteins, with a structural and dynamic continuum, is an important concept in biology, but these insights have yet to be applied in designing biomaterials. Dynamic assemblies of supramolecular phosphoglycopeptides (sPGPs) transform a 2D cell sheet into 3D cell spheroids. A ligand-receptor interaction between a glycopeptide and a phosphopeptide produces sPGPs that form nanoparticles, which transform into nanofibrils upon partial enzymatic dephosphorylation. The assemblies form dynamically and hierarchically in situ on the cell surface, and interact with the extracellular matrix molecules and effectively abolish contact inhibition of locomotion (CIL) of the cells. Integrating molecular recognition, catalysis, and assembly, these active assemblies act as a dynamic continuum to disrupt CIL, thus illustrating a new kind of biomaterial for regulating cell behavior.


Assuntos
Glicopeptídeos/metabolismo , Esferoides Celulares/química , Linhagem Celular , Fluorescência , Glicopeptídeos/química , Humanos , Substâncias Macromoleculares/química , Substâncias Macromoleculares/metabolismo , Estrutura Molecular , Esferoides Celulares/metabolismo
17.
J Am Chem Soc ; 138(49): 16046-16055, 2016 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-27960313

RESUMO

Targeting organelles by modulating the redox potential of mitochondria is a promising approach to kill cancer cells that minimizes acquired drug resistance. However, it lacks selectivity because mitochondria perform essential functions for (almost) all cells. We show that enzyme-instructed self-assembly (EISA), a bioinspired molecular process, selectively generates the assemblies of redox modulators (e.g., triphenyl phosphinium (TPP)) in the pericellular space of cancer cells for uptake, which allows selectively targeting the mitochondria of cancer cells. The attachment of TPP to a pair of enantiomeric, phosphorylated tetrapeptides produces the precursors (L-1P or D-1P) that form oligomers. Upon dephosphorylation catalyzed by ectophosphatases (e.g., alkaline phosphatase (ALP)) overexpressed on cancer cells (e.g., Saos2), the oligomers self-assemble to form nanoscale assemblies only on the surface of the cancer cells. The cancer cells thus uptake these assemblies of TPP via endocytosis, mainly via a caveolae/raft-dependent pathway. Inside the cells, the assemblies of TPP-peptide conjugates escape from the lysosome, induce dysfunction of mitochondria to release cytochrome c, and result in cell death, while the controls (i.e., omitting TPP motif, inhibiting ALP, or removing phosphate trigger) hardly kill the Saos2 cells. Most importantly, the repeated stimulation of the cancers by the precursors, unexpectedly, sensitizes the cancer cells to the precursors. As the first example of the integration of subcellular targeting with cell targeting, this study validates the spatial control of the assemblies of nonspecific cytotoxic agents by EISA as a promising molecular process for selectively killing cancer cells without inducing acquired drug resistance.


Assuntos
Fosfatase Alcalina/metabolismo , Mitocôndrias/efeitos dos fármacos , Organofosfatos/farmacologia , Biocatálise , Morte Celular/efeitos dos fármacos , Linhagem Celular , Humanos , Mitocôndrias/metabolismo , Organofosfatos/química , Fosforilação/efeitos dos fármacos
18.
J Am Chem Soc ; 138(34): 10758-61, 2016 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-27529637

RESUMO

We report that phosphotyrosine-cholesterol conjugates effectively and selectively kill cancer cells, including platinum-resistant ovarian cancer cells. The conjugate increases the degree of noncovalent oligomerization upon enzymatic dephosphorylation in aqueous buffer. This enzymatic conversion also results in the assembly of the cholesterol conjugates inside and outside cells and leads to cell death. Preliminary mechanistic studies suggest that the formed assemblies of the conjugates not only interact with actin filaments and microtubules but also affect lipid rafts. As the first report of multifaceted supramolecular assemblies of cholesterol conjugates against cancer cells, this work illustrates the integration of enzyme catalysis and self-assembly of essential biological small molecules on and inside cancer cells as a promising strategy for developing multifunctional therapeutics to treat drug-resistant cancers.


Assuntos
Antineoplásicos/química , Antineoplásicos/farmacologia , Colesterol/química , Colesterol/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Enzimas/metabolismo , Neoplasias Ovarianas/patologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Feminino , Humanos , Fosfotirosina/química
19.
Adv Healthc Mater ; : e2304574, 2024 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-38739747

RESUMO

Increasing the potency, quality, and durability of vaccines represents a major public health challenge. A critical parameter that shapes vaccine immunity is the spatiotemporal context in which immune cells interact with antigen and adjuvant. While various material-based strategies demonstrate that extended antigen release enhances both cellular and humoral immunity, the effect of adjuvant kinetics on vaccine-mediated immunity remains incompletely understood. Here, a previously characterized mesoporous silica rod (MPS) biomaterial vaccine is used to develop a facile, electrostatics-driven approach to tune in vivo kinetics of the TLR9 agonist cytosine phosphoguanosine oligodeoxynucleotide (CpG). It is demonstrated that rapid release of CpG from MPS vaccines, mediated by alterations in MPS chemistry that tune surface charge, generates potent cytotoxic T cell responses and robust, T helper type 1 (Th1)-skewed IgG2a/c antibody titers. Immunophenotyping of lymphoid organs after MPS vaccination with slow or fast CpG release kinetics suggests that differential engagement of migratory dendritic cells and natural killer cells may contribute to the more potent responses observed with rapid adjuvant release. Taken together, these findings suggest that vaccine approaches that pair sustained release of antigen with rapid release of adjuvants with similar characteristics to CpG may drive particularly potent Th1 responses.

20.
J Med Chem ; 66(14): 10027-10035, 2023 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-37459116

RESUMO

Cancer is a major public health concern requiring novel treatment approaches. Enzyme-instructed self-assembly (EISA) provides a unique approach for selectively inhibiting cancer cells. However, the structure and activity correlation of EISA remains to be explored. This study investigates new EISA substrates of alkaline phosphatase (ALP) to hinder ovarian cancer cells. Analogues 2-8 were synthesized by modifying the amino acid residues of a potent EISA substrate 1 that effectively inhibits the growth of OVSAHO, a high-grade serous ovarian cancer (HGSOC) cell line. The efficacy of 2-8 against OVSAHO was assessed, along with the combination of substrate 1 with clinically used drugs. The results reveal that substrate 1 displays the highest cytotoxicity against OVSAHO cells, with an IC50 of around 8 µM. However, there was limited synergism observed between substrate 1 and the tested clinically used drugs. These findings indicate that EISA likely operates through a distinct mechanism that necessitates further elucidation.


Assuntos
Fosfatase Alcalina , Neoplasias Ovarianas , Feminino , Humanos , Fosfatase Alcalina/metabolismo , Peptídeos/farmacologia , Peptídeos/química , Neoplasias Ovarianas/tratamento farmacológico , Linhagem Celular , Linhagem Celular Tumoral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA