Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
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.

2.
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
3.
Cell Mol Bioeng ; 15(6): 535-551, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36531864

RESUMO

Introduction: Tumor and immune cells interact through a variety of cell-surface proteins that can either restrain or promote tumor progression. The impacts of cytotoxic chemotherapy dose and delivery route on this interaction profile remain incompletely understood, and could support the development of more effective combination therapies for cancer treatment. Methods and Results: Here, we found that exposure to the anthracycline doxorubicin altered the expression of numerous immune-interacting markers (MHC-I, PD-L1, PD-L2, CD47, Fas, and calreticulin) on live melanoma, breast cancer, and leukemia cells in a dose-dependent manner in vitro. Notably, an intermediate dose best induced immunogenic cell death and the expression of immune-activating markers without maximizing expression of markers associated with immune suppression. Bone marrow-derived dendritic cells exposed to ovalbumin-expressing melanoma treated with intermediate doxorubicin dose became activated and best presented tumor antigen. In a murine melanoma model, both the doxorubicin dose and delivery location (systemic infusion versus local administration) affected the expression of these markers on live tumor cells. Particularly, local release of doxorubicin from a hydrogel increased calreticulin expression on tumor cells without inducing immune-suppressive markers, in a manner dependent on the loaded dose. Doxorubicin exposure also altered the expression of immune-interacting markers in patient-derived melanoma cells. Conclusions: Together, these results illustrate how standard-of-care chemotherapy, when administered in various manners, can lead to distinct expression of immunogenic markers on cancer cells. These findings may inform development of chemo-immunotherapy combinations for cancer treatment. Supplementary Information: The online version contains supplementary material available at 10.1007/s12195-022-00742-y.

4.
Pept Sci (Hoboken) ; 112(1)2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32104754

RESUMO

Despite multiple new-drug approvals in recent years, prostate cancer remains a global health challenge because of the prostate cancers are resistant to androgen deprivation therapy. Here we show that a small D-phosphopeptide undergoes prostatic acid phosphatase (PAP)-instructed self-assembly for inhibiting castration-resistant prostate cancer (CRPC) cells. Specifically, the installation of phosphate at the C-terminal of a D-tripeptide results in the D-phosphopeptide. Dephosphorylating the D-phosphopeptide by PAP forms uniform nanofibers that inhibit VCaP, a castration-resistant prostate cancer cell. A non-hydrolyzable phosphate analogue of the D-phosphopeptide, which shares similar self-assembling properties with the D-phosphopeptide, confirms that PAP-instructed assembly is critical for the inhibition of VCaP. This work, for the first time, demonstrates PAP-instructed self-assembly of peptides for selective inhibiting castration-resistant prostate cancer (CRPC) cells.

5.
Chem ; 5(9): 2442-2449, 2019 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-31552305

RESUMO

Alkaline phosphatases (ALP) contribute to immunosuppression in solid tumors, but they, unfortunately, are "undruggable". Here we report enzyme-instructed assembly of peptides for selectively inhibiting the tumors that overexpress ALP. We developed a precursor with two parts; an amphiphilic, self-assembling peptides joined to a hydrophilic block (i.e., tyrosine phosphate). ALP, overexpressed on and in osteosarcoma cancer cells (e.g., Saos-2), cleaves the phosphates from the tyrosine residue of the precursor and triggers the self-assembly of the resulting peptides. Being selectively formed on and inside the cancer cells, the peptide assemblies induce the cancer cell death and efficiently inhibit the tumor growth in an orthotopic osteosarcoma mice model without harming normal organs. Accordingly, the peptide assemblies significantly improve the survival ratio of metastatic tumor bearing mice. Without relying on inhibiting ALP, this approach integrates enzyme reaction and molecular self-assembly for generating peptide fibrils as potential anticancer therapeutics.

6.
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
7.
Theranostics ; 9(11): 3213-3222, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31244950

RESUMO

Using non-covalent interactions between nucleic acids (DNA, siRNA, miRNA, and mRNA) with peptides or nucleopeptides is a promising strategy to construct supramolecular assemblies for gene delivery and therapy. Comparing to conventional strategies for gene delivery, the assemblies of peptides or nucleopeptides provide several unique advantages: i) reversible interactions between the assemblies and the nucleic acids; ii) minimal immunogenicity; iii) biocompatibility. This field has advanced considerably in recent years so that it is worth summarizing the recent progresses and future challenges. In this review, we introduce the development of assemblies of peptides or nucleopeptides for applications in gene delivery and related fields. After introducing the promises of gene therapy and the current strategies for the delivery, we discuss the unique advantage of using peptide assemblies for gene delivery. Then we describe several representative strategies for gene delivery by the assemblies of peptides or nucleopeptides. Finally, we discuss the key factors for designing such assemblies for gene delivery, and speculate future directions and challenges in the field, particularly the rational design and the spatiotemporally controlled release in live cells.


Assuntos
DNA/metabolismo , Técnicas de Transferência de Genes , Substâncias Macromoleculares/metabolismo , Nucleoproteínas/metabolismo , Peptídeos/metabolismo , RNA/metabolismo , Terapia Genética/métodos , Terapia Genética/tendências , Humanos , Ligação Proteica
8.
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
9.
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
10.
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
11.
Mol Cancer Res ; 17(4): 907-917, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30552234

RESUMO

Acquired drug resistance remains a challenge in chemotherapy. Here we show enzymatic, in situ assembling of cholesterol derivatives to act as polypharmaceuticals for selectively inducing death of cancer cells via multiple pathways and without inducing acquired drug resistance. A conjugate of tyrosine and cholesterol (TC), formed by enzyme-catalyzed dephosphorylation of phosphorylate TC, self-assembles selectively on or in cancer cells. Acting as polypharmaceuticals, the assemblies of TC augment lipid rafts, aggregate extrinsic cell death receptors (e.g., DR5, CD95, or TRAILR), modulate the expression of oncoproteins (e.g., Src and Akt), disrupt the dynamics of cytoskeletons (e.g., actin filaments or microtubules), induce endoplasmic reticulum stress, and increase the production of reactive oxygen species, thus resulting in cell death and preventing acquired drug resistance. Moreover, the assemblies inhibit the growth of platinum-resistant ovarian cancer tumor in a murine model. This work illustrates the use of instructed assembly (iA) in cellular environment to form polypharmaceuticals in situ that not only interact with multiple proteins, but also modulate membrane dynamics for developing novel anticancer therapeutics. IMPLICATIONS: As a multifaceted strategy for controlling cancer cell death, iA minimized acquired resistance of cancer cells, which is a new strategy to amplify the genetic difference between cancer and normal cells and provides a promise for overcoming drug resistance in cancer therapy.Visual Overview: http://mcr.aacrjournals.org/content/molcanres/17/4/907/F1.large.jpg.


Assuntos
Colesterol/análogos & derivados , Colesterol/farmacologia , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Actinas/metabolismo , Animais , Antineoplásicos/farmacologia , Morte Celular/fisiologia , Linhagem Celular Tumoral , Cisplatino/farmacologia , Doxorrubicina/farmacologia , Resistencia a Medicamentos Antineoplásicos , Estresse do Retículo Endoplasmático , Células HeLa , Células Hep G2 , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Dinâmica Mitocondrial/efeitos dos fármacos , Neoplasias/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Quinases da Família src/metabolismo
12.
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
13.
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
14.
Chem Asian J ; 13(22): 3464-3468, 2018 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-29897657

RESUMO

Nanofibers of short peptides are emerging as a promising type of agents for inhibiting cancer cells. But the proteolysis of peptides decreases the anticancer efficacy of the peptide nanofibers. Here we show that decreasing the activity of proteasomes enhance the activity of peptide nanofibers for inhibiting cancer cells. Based on the structure of galactin-3, we designed a heptapeptide, which self-assembles to form nanofibers. The nanofibers of the heptapeptide exhibit moderate cytotoxicity to three representative cancer cell lines (HeLa, MCF-7, and HepG2), largely due to the proteolysis of the peptides. Using a clinically approved proteasome inhibitor, bortezomib, to treat the cancer cells significantly decreases the proteolysis of the peptides and enhances the activity of the peptide nanofibers for inhibiting the cancer cells. This work illustrates a promising approach for enhancing the anticancer efficacy of peptide nanofibers by modulating intracellular protein degradation machinery, as well as provides insights for understanding the cytotoxicity of aberrant protein or peptide aggregates in complicated cellular environment.


Assuntos
Nanofibras/química , Peptídeos/química , Sequência de Aminoácidos , Sobrevivência Celular/efeitos dos fármacos , Galectina 3/química , Células HeLa , Células Hep G2 , Humanos , Hidrogéis/química , Células MCF-7 , Peptídeos/metabolismo , Peptídeos/farmacologia , Proteólise
15.
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
16.
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
17.
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
18.
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
19.
Adv Drug Deliv Rev ; 110-111: 102-111, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27102943

RESUMO

Nanostructures formed by peptides that self-assemble in water through non-covalent interactions have attracted considerable attention because peptides possess several unique advantages, such as modular design and easiness of synthesis, convenient modification with known functional motifs, good biocompatibility, low immunogenicity and toxicity, inherent biodegradability, and fast responses to a wide range of external stimuli. After about two decades of development, peptide-based supramolecular nanostructures have already shown great potentials in the fields of biomedicine. Among a range of biomedical applications, using such nanostructures for cancer therapy has attracted increased interests since cancer remains the major threat for human health. Comparing with L-peptides, nanostructures containing peptides made of D-amino acid (i.e., D-peptides) bear a unique advantage, biostability (i.e., resistance towards most of endogenous enzymes). The exploration of nanostructures containing D-amino acids, especially their biomedical applications, is still in its infancy. Herein we review the recent progress of D-amino acid-containing supramolecular nanofibers as an emerging class of biomaterials that exhibit unique features for the development of cancer therapeutics. In addition, we give a brief perspective about the challenges and promises in this research direction.


Assuntos
Aminoácidos/química , Nanofibras/química , Nanofibras/uso terapêutico , Neoplasias/tratamento farmacológico , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/química , Antineoplásicos/uso terapêutico , Materiais Biocompatíveis/química , Materiais Biocompatíveis/uso terapêutico , Portadores de Fármacos/química , Humanos , Neoplasias/metabolismo , Peptídeos/química , Peptídeos/uso terapêutico
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA