Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Ther ; 30(2): 672-687, 2022 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-34274535

RESUMO

Triple-negative breast cancer (TNBC) has a high propensity for organ-specific metastasis. However, the underlying mechanisms are not well understood. Here we show that the primary TNBC tumor-derived C-X-C motif chemokines 1/2/8 (CXCL1/2/8) stimulate lung-resident fibroblasts to produce the C-C motif chemokines 2/7 (CCL2/7), which, in turn, activate cholesterol synthesis in lung-colonizing TNBC cells and induce angiogenesis at lung metastatic sites. Inhibiting cholesterol synthesis in lung-colonizing breast tumor cells by pulmonary administration of simvastatin-carrying HER3-targeting nanoparticles reduces angiogenesis and growth of lung metastases in a syngeneic TNBC mouse model. Our findings reveal a novel, chemokine-regulated mechanism for the cholesterol synthesis pathway and a critical role of metastatic site-specific cholesterol synthesis in the pulmonary tropism of TNBC metastasis. The study has implications for the unresolved epidemiological observation that use of cholesterol-lowering drugs has no effect on breast cancer incidence but can unexpectedly reduce breast cancer mortality, suggesting interventions of cholesterol synthesis in lung metastases as an effective treatment to improve survival in individuals with TNBC.


Assuntos
Neoplasias de Mama Triplo Negativas , Animais , Linhagem Celular Tumoral , Quimiocinas , Humanos , Pulmão/metabolismo , Camundongos , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/genética , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/genética
2.
Nucleic Acids Res ; 47(21): 11020-11043, 2019 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-31617560

RESUMO

RNA interference represents a potent intervention for cancer treatment but requires a robust delivery agent for transporting gene-modulating molecules, such as small interfering RNAs (siRNAs). Although numerous molecular approaches for siRNA delivery are adequate in vitro, delivery to therapeutic targets in vivo is limited by payload integrity, cell targeting, efficient cell uptake, and membrane penetration. We constructed nonviral biomaterials to transport small nucleic acids to cell targets, including tumor cells, on the basis of the self-assembling and cell-penetrating activities of the adenovirus capsid penton base. Our recombinant penton base chimera contains polypeptide domains designed for noncovalent assembly with anionic molecules and tumor homing. Here, structural modeling, molecular dynamics simulations, and functional assays suggest that it forms pentameric units resembling viral capsomeres that assemble into larger capsid-like structures when combined with siRNA cargo. Pentamerization forms a barrel lined with charged residues mediating pH-responsive dissociation and exposing masked domains, providing insight on the endosomolytic mechanism. The therapeutic impact was examined on tumors expressing high levels of HER3/ErbB3 that are resistant to clinical inhibitors. Our findings suggest that our construct may utilize ligand mimicry to avoid host attack and target the siRNA to HER3+ tumors by forming multivalent capsid-like structures.


Assuntos
Portadores de Fármacos/uso terapêutico , Nanopartículas/uso terapêutico , RNA Interferente Pequeno/farmacologia , Receptor ErbB-3/antagonistas & inibidores , Proteínas Recombinantes/uso terapêutico , Animais , Proteínas do Capsídeo/química , Linhagem Celular Tumoral , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Neuregulina-1/química , Interferência de RNA
3.
J Allergy Clin Immunol ; 142(6): 1808-1817.e3, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-29522849

RESUMO

BACKGROUND: Altered epithelial physical and functional barrier properties along with TH1/TH2 immune dysregulation are features of allergic asthma. Regulation of junction proteins to improve barrier function of airway epithelial cells has the potential for alleviation of allergic airway inflammation. OBJECTIVE: We sought to determine the immunomodulatory effect of knob protein of the adenoviral capsid on allergic asthma and to investigate its mechanism of action on airway epithelial junction proteins and barrier function. METHODS: Airway inflammation, including junction protein expression, was evaluated in allergen-challenged mice with and without treatment with knob. Human bronchial epithelial cells were exposed to knob, and its effects on expression of junction proteins and barrier integrity were determined. RESULTS: Administration of knob to allergen-challenged mice suppressed airway inflammation (eosinophilia, airway hyperresponsiveness, and IL-5 levels) and prevented allergen-induced loss of airway epithelial occludin and E-cadherin expression. Additionally, knob decreased expression of TH2-promoting inflammatory mediators, specifically IL-33, by murine lung epithelial cells. At a cellular level, treatment of human bronchial epithelial cells with knob activated c-Jun N-terminal kinase, increased expression of occludin and E-cadherin, and enhanced epithelial barrier integrity. CONCLUSION: Increased expression of junction proteins mediated by knob leading to enhanced epithelial barrier function might mitigate the allergen-induced airway inflammatory response, including asthma.


Assuntos
Proteínas do Capsídeo/farmacologia , Proteínas do Capsídeo/uso terapêutico , Células Epiteliais/efeitos dos fármacos , Adenoviridae , Idoso , Animais , Brônquios/citologia , Líquido da Lavagem Broncoalveolar/imunologia , Caderinas/metabolismo , Linhagem Celular , Citocinas/imunologia , Eosinofilia/imunologia , Células Epiteliais/metabolismo , Feminino , Humanos , Masculino , Camundongos Endogâmicos BALB C , Pessoa de Meia-Idade , Ocludina/metabolismo , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Hipersensibilidade Respiratória/tratamento farmacológico , Hipersensibilidade Respiratória/imunologia
4.
Cancers (Basel) ; 15(12)2023 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-37370850

RESUMO

Macromolecular therapeutics such as nucleic acids, peptides, and proteins have the potential to overcome treatment barriers for cancer. For example, nucleic acid or peptide biologics may offer an alternative strategy for attacking otherwise undruggable therapeutic targets such as transcription factors and similar oncologic drivers. Delivery of biological therapeutics into tumor cells requires a robust system of cell penetration to access therapeutic targets within the cell interior. A highly effective means of accomplishing this may be borrowed from cell-penetrating pathogens such as viruses. In particular, the cell entry function of the adenovirus penton base capsid protein has been effective at penetrating tumor cells for the intracellular deposition of macromolecular therapies and membrane-impermeable drugs. Here, we provide an overview describing the evolution of tumor-targeted penton-base-derived nano-capsids as a framework for discussing the requirements for overcoming key barriers to macromolecular delivery. The development and pre-clinical testing of these proteins for therapeutic delivery has begun to also uncover the elusive mechanism underlying the membrane-penetrating function of the penton base. An understanding of this mechanism may unlock the potential for macromolecular therapeutics to be effectively delivered into cancer cells and to provide a treatment option for tumors resisting current clinical therapies.

5.
Proc Natl Acad Sci U S A ; 106(15): 6105-10, 2009 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-19342490

RESUMO

Sulfonated gallium(III) corroles are intensely fluorescent macrocyclic compounds that spontaneously assemble with carrier proteins to undergo cell entry. We report in vivo imaging and therapeutic efficacy of a tumor-targeted corrole noncovalently assembled with a heregulin-modified protein directed at the human epidermal growth factor receptor (HER). Systemic delivery of this protein-corrole complex results in tumor accumulation, which can be visualized in vivo owing to intensely red corrole fluorescence. Targeted delivery in vivo leads to tumor cell death while normal tissue is spared. These findings contrast with the effects of doxorubicin, which can elicit cardiac damage during therapy and required direct intratumoral injection to yield similar levels of tumor shrinkage compared with the systemically delivered corrole. The targeted complex ablated tumors at >5 times a lower dose than untargeted systemic doxorubicin, and the corrole did not damage heart tissue. Complexes remained intact in serum and the carrier protein elicited no detectable immunogenicity. The sulfonated gallium(III) corrole functions both for tumor detection and intervention with safety and targeting advantages over standard chemotherapeutic agents.


Assuntos
Metaloporfirinas , Neoplasias/diagnóstico , Animais , Anticorpos/imunologia , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Neoplasias/imunologia , Neoplasias/metabolismo , Receptor ErbB-2/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Cancer Lett ; 534: 215613, 2022 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-35276290

RESUMO

Signal transducer and activator of transcription (Stat)3 is a valid anticancer therapeutic target. We have discovered a highly potent chemotype that amplifies the Stat3-inhibitory activity of lead compounds to levels previously unseen. The azetidine-based compounds, including H172 (9f) and H182, irreversibly bind to Stat3 and selectively inhibit Stat3 activity (IC50 0.38-0.98 µM) over Stat1 or Stat5 (IC50 > 15.8 µM) in vitro. Mass spectrometry detected the Stat3 cysteine peptides covalently bound to the azetidine compounds, and the key residues, Cys426 and Cys468, essential for the high potency inhibition, were confirmed by site-directed mutagenesis. In triple-negative breast cancer (TNBC) models, treatment with the azetidine compounds inhibited constitutive and ligand-induced Stat3 signaling, and induced loss of viable cells and tumor cell death, compared to no effect on the induction of Janus kinase (JAK)2, Src, epidermal growth factor receptor (EGFR), and other proteins, or weak effects on cells that do not harbor aberrantly-active Stat3. H120 (8e) and H182 as a single agent inhibited growth of TNBC xenografts, and H278 (hydrochloric acid salt of H182) in combination with radiation completely blocked mouse TNBC growth and improved survival in syngeneic models. We identify potent azetidine-based, selective, irreversible Stat3 inhibitors that inhibit TNBC growth in vivo.


Assuntos
Azetidinas , Neoplasias de Mama Triplo Negativas , Animais , Apoptose , Azetidinas/farmacologia , Linhagem Celular Tumoral , Humanos , Camundongos , Fosforilação , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/genética
7.
Mol Pharm ; 8(6): 2233-43, 2011 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-21981771

RESUMO

HerGa is a self-assembled tumor-targeted particle that bears both tumor detection and elimination activities in a single, two-component complex (Agadjanian et al. Proc. Natl. Acad. Sci. U.S.A.2009, 106, 6105-6110). Given its multifunctionality, HerGa (composed of the fluorescent cytotoxic corrole macrocycle, S2Ga, noncovalently bound to the tumor-targeted cell penetration protein, HerPBK10) has the potential for high clinical impact, but its mechanism of cell killing remains to be elucidated, and hence is the focus of the present study. Here we show that HerGa requires HerPBK10-mediated cell entry to induce toxicity. HerGa (but not HerPBK10 or S2Ga alone) induced mitochondrial membrane potential disruption and superoxide elevation, which were both prevented by endosomolytic-deficient mutants, indicating that cytosolic exposure is necessary for corrole-mediated cell death. A novel property discovered here is that corrole fluorescence lifetime acts as a pH indicator, broadcasting the intracellular microenvironmental pH during uptake in live cells. This feature in combination with two-photon imaging shows that HerGa undergoes early endosome escape during uptake, avoiding compartments of pH < 6.5. Cytoskeletal disruption accompanied HerGa-mediated mitochondrial changes whereas oxygen scavenging reduced both events. Paclitaxel treatment indicated that HerGa uptake requires dynamic microtubules. Unexpectedly, low pH is insufficient to induce release of the corrole from HerPBK10. Altogether, these studies identify a mechanistic pathway in which early endosomal escape enables HerGa-induced superoxide generation leading to cytoskeletal and mitochondrial damage, thus triggering downstream cell death.


Assuntos
Sistemas de Liberação de Medicamentos , Imunotoxinas/toxicidade , Porfirinas/toxicidade , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Humanos , Concentração de Íons de Hidrogênio , Microscopia Confocal , Modelos Biológicos , Neoplasias/patologia , Paclitaxel/farmacologia
8.
J Med Chem ; 64(1): 695-710, 2021 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-33352047

RESUMO

We optimized our previously reported proline-based STAT3 inhibitors into an exciting new series of (R)-azetidine-2-carboxamide analogues that have sub-micromolar potencies. 5a, 5o, and 8i have STAT3-inhibitory potencies (IC50) of 0.55, 0.38, and 0.34 µM, respectively, compared to potencies greater than 18 µM against STAT1 or STAT5 activity. Further modifications derived analogues, including 7e, 7f, 7g, and 9k, that addressed cell membrane permeability and other physicochemical issues. Isothermal titration calorimetry analysis confirmed high-affinity binding to STAT3, with KD of 880 nM (7g) and 960 nM (9k). 7g and 9k inhibited constitutive STAT3 phosphorylation and DNA-binding activity in human breast cancer, MDA-MB-231 or MDA-MB-468 cells. Furthermore, treatment of breast cancer cells with 7e, 7f, 7g, or 9k inhibited viable cells, with an EC50 of 0.9-1.9 µM, cell growth, and colony survival, and induced apoptosis while having relatively weaker effects on normal breast epithelial, MCF-10A or breast cancer, MCF-7 cells that do not harbor constitutively active STAT3.


Assuntos
Azetidinas/química , Fator de Transcrição STAT3/antagonistas & inibidores , Amidas/química , Apoptose/efeitos dos fármacos , Azetidinas/metabolismo , Azetidinas/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , DNA/química , DNA/metabolismo , Avaliação Pré-Clínica de Medicamentos , Humanos , Concentração Inibidora 50 , Fosforilação/efeitos dos fármacos , Ligação Proteica , Fator de Transcrição STAT3/metabolismo , Relação Estrutura-Atividade
9.
JCI Insight ; 52019 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-31393853

RESUMO

Idiopathic pulmonary fibrosis (IPF) is a chronic and fatal lung disease. A maladaptive epithelium due to chronic injury is a prominent feature and contributor to pathogenic cellular communication in IPF. Recent data highlight the concept of a "reprogrammed" lung epithelium as critical in the development of lung fibrosis. Extracellular vesicles (EVs) are potent mediator of cellular crosstalk, and recent evidence supports their role in lung pathologies such as IPF. Here, we demonstrate that syndecan-1 is overexpressed by the epithelium in the lungs of IPF patients and in murine models after bleomycin injury. Moreover, we find that syndecan-1 is a pro-fibrotic signal that alters alveolar type II (ATII) cell phenotypes by augmenting TGFß and Wnt signaling among other pro-fibrotic pathways. Importantly, we demonstrate that syndecan-1 controls the packaging of several anti-fibrotic microRNAs into EVs that have broad effects over several fibrogenic signaling networks as a mechanism of regulating epithelial plasticity and pulmonary fibrosis. Collectively, our work reveals new insight into how EVs orchestrate cellular signals that promote lung fibrosis and demonstrate the importance of syndecan-1 in coordinating these programs.


Assuntos
Células Epiteliais Alveolares/metabolismo , Vesículas Extracelulares/metabolismo , Fibrose Pulmonar Idiopática/metabolismo , Sindecana-1/metabolismo , Células Epiteliais Alveolares/patologia , Animais , Bleomicina/efeitos adversos , Linhagem Celular , Modelos Animais de Doenças , Vesículas Extracelulares/patologia , Feminino , Humanos , Fibrose Pulmonar Idiopática/induzido quimicamente , Fibrose Pulmonar Idiopática/patologia , Pulmão/patologia , Lesão Pulmonar/induzido quimicamente , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , MicroRNAs/genética , MicroRNAs/metabolismo , Transdução de Sinais , Sindecana-1/genética , Transcriptoma , Fator de Crescimento Transformador beta/metabolismo
10.
J Inorg Biochem ; 102(3): 446-57, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18180041

RESUMO

The interactions of transferrin (Tf) with the water soluble corrole 1 and with its gallium (1-Ga) and manganese (1-Mn) complexes were studied to establish the possible utilization of corrole-transferrin conjugates for targeting these corroles to cells that express the transferrin receptor. The protein, in both its iron-free apo form (apoTf) and the iron-bound holo form (holoTf), was found to spontaneously bind all three derivatives. This conclusion was reached from titrations followed by several spectroscopic methods and dilution experiments measured by fluorescence. The such elucidated very small dissociation constant of 2 x 10(-7) M and 3 x 10(-8) M for 1-Ga with apoTf and holoTf, respectively and <10(-9) M for 1 with both protein forms are clearly relevant for the physiological concentration of transferrin in serum.


Assuntos
Porfirinas/química , Porfirinas/metabolismo , Transferrina/química , Transferrina/metabolismo , Eletroforese em Gel de Poliacrilamida , Gálio/química , Gálio/metabolismo , Magnésio/química , Magnésio/metabolismo , Estrutura Molecular , Ligação Proteica , Análise de Regressão
11.
J Control Release ; 271: 127-138, 2018 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-29288681

RESUMO

Resistance to anti-tumor therapeutics is an important clinical problem. Tumor-targeted therapies currently used in the clinic are derived from antibodies or small molecules that mitigate growth factor activity. These have improved therapeutic efficacy and safety compared to traditional treatment modalities but resistance arises in the majority of clinical cases. Targeting such resistance could improve tumor abatement and patient survival. A growing number of such tumors are characterized by prominent expression of the human epidermal growth factor receptor 3 (HER3) on the cell surface. This study presents a "Trojan-Horse" approach to combating these tumors by using a receptor-targeted biocarrier that exploits the HER3 cell surface protein as a portal to sneak therapeutics into tumor cells by mimicking an essential ligand. The biocarrier used here combines several functions within a single fusion protein for mediating targeted cell penetration and non-covalent self-assembly with therapeutic cargo, forming HER3-homing nanobiologics. Importantly, we demonstrate here that these nanobiologics are therapeutically effective in several scenarios of resistance to clinically approved targeted inhibitors of the human EGF receptor family. We also show that such inhibitors heighten efficacy of our nanobiologics on naïve tumors by augmenting HER3 expression. This approach takes advantage of a current clinical problem (i.e. resistance to growth factor inhibition) and uses it to make tumors more susceptible to HER3 nanobiologic treatment. Moreover, we demonstrate a novel approach in addressing drug resistance by taking inhibitors against which resistance arises and re-introducing these as adjuvants, sensitizing tumors to the HER3 nanobiologics described here.


Assuntos
Antineoplásicos/administração & dosagem , Produtos Biológicos/administração & dosagem , Portadores de Fármacos/administração & dosagem , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Nanopartículas/administração & dosagem , Peptídeos/administração & dosagem , Receptor ErbB-3/metabolismo , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Feminino , Humanos , Camundongos , Neoplasias/tratamento farmacológico
12.
Clin Breast Cancer ; 15(6): 448-457.e2, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26248960

RESUMO

BACKGROUND: We investigated the effect of basal protein expression on trastuzamab response in patients with HER2-positive (HER2(+)) breast cancer who received trastuzamab (T) and in HER2(+) breast cancer cell lines. PATIENTS AND METHODS: Expression of cytokeratin (CK) 5/6, CK14, and epidermal growth factor receptor (EGFR) was evaluated after immunohistochemical staining in paraffin-embedded tissue of 97 patients with stage I to III HER2(+) breast cancer treated with chemotherapy/T. Groups with and without basal protein expression were compared with respect to clinicopathologic parameters and survival. We treated 4 cell lines (2 basal-HER2 [HCC1569, HCC1954] and 2 nonbasal HER2 [BT474, SKBR3]) each with vehicle, T 20 µg/mL, paclitaxel 0.01 µM (P), and T with P (T + P). Cell viability was assessed and HER2 pathway suppression was compared between groups using immunoblot analysis. Mammosphere formation was used to assess breast cancer stem cell properties. RESULTS: EGFR expression was significantly associated with cancer-specific survival (CSS) (P = .05). CK5/6 expression strongly correlated with overall and disease-free survival, and CSS (P = .03, P = .04, and P = .03, respectively). Statistical significance was maintained for EGFR and CK5/6 after adjustment for covariates. CK14 was not associated with survival. All cell lines expressed similar levels of HER2. T and P alone inhibited proliferation of nonbasal cell lines; T + P had an additive cytotoxic effect. Basal cells were resistant to T, P inhibited proliferation, but T + P had no additive cytotoxic effect on cell growth in basal cells. Immunoblot analysis showed a significant decrease in phosphorylated Akt levels after treatment with T or T + P in nonbasal cells but not in basal cells. Akt blockade suppressed growth of basal and nonbasal HER2(+) cells. Furthermore, basal HER2 cell lines had increased mammosphere formation, which suggests increased stem cell properties compared with nonbasal HER2 cell lines. CONCLUSION: CK5/6 and EGFR expression are predictive of worse prognosis in HER2(+) breast cancer patients treated with T. Basal HER2 breast cancer cell lines are resistant to trastuzamab, which is mediated through the Akt pathway; AKT inhibition abrogates this resistance. Basal HER2 cell lines also have increased stem cell properties, which might play a role in the resistance pathway.


Assuntos
Biomarcadores Tumorais/análise , Neoplasias da Mama/patologia , Resistencia a Medicamentos Antineoplásicos/fisiologia , Receptores ErbB/biossíntese , Queratinas/biossíntese , Adulto , Idoso , Idoso de 80 Anos ou mais , Antineoplásicos/uso terapêutico , Neoplasias da Mama/metabolismo , Neoplasias da Mama/mortalidade , Intervalo Livre de Doença , Receptores ErbB/análise , Feminino , Humanos , Immunoblotting , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Queratinas/análise , Pessoa de Meia-Idade , Invasividade Neoplásica , Fenótipo , Modelos de Riscos Proporcionais , Receptor ErbB-2/antagonistas & inibidores , Trastuzumab/uso terapêutico , Resultado do Tratamento
13.
J Control Release ; 217: 92-101, 2015 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-26334483

RESUMO

Water-soluble corroles with inherent fluorescence can form stable self-assemblies with tumor-targeted cell penetration proteins, and have been explored as agents for optical imaging and photosensitization of tumors in pre-clinical studies. However, the limited tissue-depth of excitation wavelengths limits their clinical applicability. To examine their utility in more clinically-relevant imaging and therapeutic modalities, here we have explored the use of corroles as contrast enhancing agents for magnetic resonance imaging (MRI), and evaluated their potential for tumor-selective delivery when encapsulated by a tumor-targeted polypeptide. We have found that a manganese-metallated corrole exhibits significant T1 relaxation shortening and MRI contrast enhancement that is blocked by particle formation in solution but yields considerable MRI contrast after tissue uptake. Cell entry but not low pH enables this. Additionally, the corrole elicited tumor-toxicity through the loss of mitochondrial membrane potential and cytoskeletal breakdown when delivered by the targeted polypeptide. The protein-corrole particle (which we call HerMn) exhibited improved therapeutic efficacy compared to current targeted therapies used in the clinic. Taken together with its tumor-preferential biodistribution, our findings indicate that HerMn can facilitate tumor-targeted toxicity after systemic delivery and tumor-selective MR imaging activatable by internalization.


Assuntos
Antineoplásicos , Meios de Contraste , Manganês , Neuregulina-1 , Porfirinas , Animais , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Meios de Contraste/farmacocinética , Meios de Contraste/farmacologia , Meios de Contraste/uso terapêutico , Feminino , Humanos , Imageamento por Ressonância Magnética , Manganês/farmacocinética , Manganês/farmacologia , Manganês/uso terapêutico , Camundongos Nus , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Neoplasias/patologia , Neuregulina-1/farmacocinética , Neuregulina-1/farmacologia , Neuregulina-1/uso terapêutico , Porfirinas/farmacocinética , Porfirinas/farmacologia , Porfirinas/uso terapêutico , Proteínas Recombinantes de Fusão/farmacocinética , Proteínas Recombinantes de Fusão/farmacologia , Proteínas Recombinantes de Fusão/uso terapêutico , Distribuição Tecidual , Carga Tumoral/efeitos dos fármacos
14.
Adv Drug Deliv Rev ; 55(11): 1485-96, 2003 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-14597142

RESUMO

Key proteins of the icosahedral-shaped adenovirus (Ad) capsid mediate infection, and interact with cellular proteins to coordinate stepwise events of cell entry that produce successful gene transfer. Infection is mediated predominantly by the penton and fiber capsid proteins. The fiber initiates cell binding while the penton binds integrin coreceptors, triggering integrin-mediated endocytosis. Penton integrin signaling precedes viral escape from the endosomal vesicle. After cell binding, the virus undergoes stepwise disassembly of the capsid, shedding proteins during cell entry. Intracellular trafficking of the remaining capsid shell is mediated by the interaction of naked particles with the cytoskeleton. The capsid translocates toward the nucleus, with the majority of capsid proteins accumulating at the nuclear periphery, while viral DNA and associated protein VII are extruded through the nuclear pore. This discussion will encompass the current knowledge on Ad cell entry and trafficking, with an emphasis on the contribution of Ad capsid proteins to these processes. A greater understanding of the highly effective Ad cell entry pathway may lend itself to the development of safer drug and gene delivery alternatives utilizing similar pathways.


Assuntos
Adenoviridae/fisiologia , Proteínas do Capsídeo/fisiologia , Endocitose/fisiologia , Adenoviridae/genética , Adenoviridae/metabolismo , Animais , Transporte Biológico , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Citoesqueleto/metabolismo , Endossomos/metabolismo , Humanos , Transfecção
15.
J Control Release ; 93(2): 129-40, 2003 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-14636719

RESUMO

To evaluate the safety of adenovirus-derived capsid proteins for ocular gene delivery, we have investigated their effects on the morphology and function of the acinar epithelial cells of the lacrimal gland. These cells are responsible for basal and stimulated release of proteins and electrolytes into ocular fluid, a process essential in maintaining the health of the ocular surface. Acinar epithelial cells from rabbit lacrimal gland were exposed to one of two adenovirus serotype 5 capsid proteins, penton or knob (the carboxy-terminal fragment of the fiber capsid protein). Sustained (16-18 h) exposure to the penton at 20 microg/ml was associated with major changes in the organization of the regulated secretory pathway and cytoskeleton. These changes included an apparent loss of mature secretory vesicles enriched in rab3D around the apical lumen as well as a depletion of apical actin. The microtubule array in penton-treated acini also exhibited bundling and disorganization. None of these effects were elicited by exposure to knob protein. Penton treatment also caused a significant (p < or = 0.05) increase and decrease in basal and carbachol-stimulated release, respectively, of bulk protein. Competition studies showed that RGD peptide partially prevented the penton-induced changes in rab3D-enriched secretory vesicles and actin filaments. These findings suggest that the adenovirus penton protein compromises normal acinar secretory compartment organization and function and that these changes are due at least partly to penton-integrin interactions.


Assuntos
Proteínas do Capsídeo/farmacologia , Células Epiteliais/metabolismo , Actinas/efeitos dos fármacos , Actinas/metabolismo , Animais , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Carbacol/farmacologia , Células Cultivadas , Sistemas de Liberação de Medicamentos/métodos , Avaliação Pré-Clínica de Medicamentos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/ultraestrutura , Feminino , Integrina alfaVbeta3/metabolismo , Aparelho Lacrimal/química , Aparelho Lacrimal/citologia , Aparelho Lacrimal/efeitos dos fármacos , Aparelho Lacrimal/metabolismo , Camundongos , Proteínas Associadas aos Microtúbulos/efeitos dos fármacos , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas Associadas aos Microtúbulos/ultraestrutura , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/farmacologia , Faloidina/análogos & derivados , Faloidina/metabolismo , Coelhos , Rodaminas/farmacologia , Fatores de Tempo , Proteínas rab3 de Ligação ao GTP/química , Proteínas rab3 de Ligação ao GTP/efeitos dos fármacos
16.
J Inorg Biochem ; 140: 39-44, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25061689

RESUMO

We are investigating the biological and biomedical imaging roles and impacts of fluorescent metallocorrole-TiO2 nanoconjugates as potential near-infrared optical contrast agents in vitro in cancer and normal cell lines. The TiO2 nanoconjugate labeled with the small molecule 2,17-bis(chlorosulfonyl)-5,10,15-tris(pentafluorophenyl)corrolato aluminum(III) (1-Al-TiO2) was prepared. The nanoparticle 1-Al-TiO2 was characterized by transmission electron microscopy (TEM) and integrating-sphere electronic absorption spectroscopy. TEM images of three different samples of TiO2 nanoparticles (bare, H2O2 etched, and 1-Al functionalized) showed similarity in shapes and sizes with an average diameter of 29nm for 1-Al-TiO2. Loading of 1-Al on the TiO2 surfaces was determined to be ca. 20-40mg 1-Al/g TiO2. Confocal fluorescence microscopy (CFM) studies of luciferase-transfected primary human glioblastoma U87-Luc cells treated with the nanoconjugate 1-Al-TiO2 as the contrast agent in various concentrations were performed. The CFM images revealed that 1-Al-TiO2 was found inside the cancer cells even at low doses (0.02-2µg/mL) and localized in the cytosol. Bioluminescence studies of the U87-Luc cells exposed to various amounts of 1-Al-TiO2 showed minimal cytotoxic effects even at higher doses (2-2000µg/mL) after 24h. A similar observation was made using primary mouse hepatocytes (PMH) treated with 1-Al-TiO2 at low doses (0.0003-3µg/mL). Longer incubation times (after 48 and 72h for U87-Luc) and higher doses (>20µg/mL 1-Al-TiO2 for U87-Luc and >3µg/mL 1-Al-TiO2 for PMH) showed decreased cell viability.


Assuntos
Nanoestruturas , Porfirinas/química , Espectroscopia de Luz Próxima ao Infravermelho/métodos , Titânio/química , Animais , Células Cultivadas , Hepatócitos/citologia , Hepatócitos/efeitos dos fármacos , Humanos , Camundongos , Microscopia Eletrônica de Transmissão , Microscopia de Fluorescência , Nanoestruturas/toxicidade
17.
Ther Deliv ; 4(2): 267-77, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23343164

RESUMO

The outer shell of the adenovirus capsid comprises three major types of protein (hexon, penton base and fiber) that perform the majority of functions facilitating the early stages of adenovirus infection. These stages include initial cell-surface binding followed by receptor-mediated endocytosis, endosomal penetration and cytosolic entry, and intracellular trafficking toward the nucleus. Numerous studies have shown that the penton base contributes to several of these steps and have supported the development of this protein into a delivery agent for therapeutic molecules. Studies revealing that the fiber and hexon bear unexpected properties of cell entry and/or nuclear homing have supported the development of these capsid proteins, as well into potential delivery vehicles. This review summarizes the findings to date of the protein-cell activities of these capsid proteins in the absence of the whole virus and their potential for therapeutic application with regard to the delivery of foreign molecules.


Assuntos
Proteínas do Capsídeo/química , Sistemas de Liberação de Medicamentos , Técnicas de Transferência de Genes , Adenoviridae/química , Animais , Humanos
18.
J Vis Exp ; (76)2013 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-23851334

RESUMO

The HER2+ tumor-targeted nanoparticle, HerDox, exhibits tumor-preferential accumulation and tumor-growth ablation in an animal model of HER2+ cancer. HerDox is formed by non-covalent self-assembly of a tumor targeted cell penetration protein with the chemotherapy agent, doxorubicin, via a small nucleic acid linker. A combination of electrophilic, intercalation, and oligomerization interactions facilitate self-assembly into round 10-20 nm particles. HerDox exhibits stability in blood as well as in extended storage at different temperatures. Systemic delivery of HerDox in tumor-bearing mice results in tumor-cell death with no detectable adverse effects to non-tumor tissue, including the heart and liver (which undergo marked damage by untargeted doxorubicin). HER2 elevation facilitates targeting to cells expressing the human epidermal growth factor receptor, hence tumors displaying elevated HER2 levels exhibit greater accumulation of HerDox compared to cells expressing lower levels, both in vitro and in vivo. Fluorescence intensity imaging combined with in situ confocal and spectral analysis has allowed us to verify in vivo tumor targeting and tumor cell penetration of HerDox after systemic delivery. Here we detail our methods for assessing tumor targeting via multimode imaging after systemic delivery.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/enzimologia , Doxorrubicina/administração & dosagem , Nanopartículas/administração & dosagem , Receptor ErbB-2/metabolismo , Animais , Antibióticos Antineoplásicos/administração & dosagem , Antibióticos Antineoplásicos/química , Antibióticos Antineoplásicos/farmacocinética , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Doxorrubicina/química , Doxorrubicina/farmacocinética , Estabilidade de Medicamentos , Feminino , Humanos , Camundongos , Camundongos Nus , Microscopia de Fluorescência/métodos , Nanopartículas/química , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Ultrasonics ; 52(7): 803-8, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22513260

RESUMO

Targeted mechanical cell stimulation has been extensively studied for a better understanding of its effect on cellular mechanotransduction signaling pathways and structures by utilizing a variety of mechanical sources. In this work, an ultrasound-driven single cell stimulation method is thus proposed, and a preliminary study is carried out by comparing the fluorescence intensities representing a change in cell membrane permeability between MDA-MB-435 human HER2+ cancer cells (∼40-50µm in diameter) and MCF-12F normal cells (∼50-60µm) in the presence of ultrasound. A 200MHz single element zinc oxide (ZnO) transducer is employed to generate ultrasound microbeam (UM) whose beamwidth and depth of focus are 9.5 and 60µm, comparable to typical cell size. The cells in tetramethyl rhodamine methyl ester (TMRM) are interrogated with 200MHz sinusoidal bursts. The number of cycles per burst is 5 and the pulse repetition frequency (PRF) is 1kHz. The temporal variation of fluorescence intensity in each cell is measured as a function of input voltage to the transducer (16, 32, and 47V), and its corresponding fluorescence images are obtained via a confocal microscope. A systematic method for visualizing UM's focus by adding Rhodamine B to the immersion medium is also proposed to enhance the precision in aiming the beam at an individual cell. Both types of cells exhibit a decrease in the intensity upon UM irradiation. In particular, normal cells show more fluorescence reduction (down to 0.7 in normalized intensity) than cancer cells (∼0.9) under the same excitation condition of the transducer. With UM being turned off, the normalized intensity level in normal cells is slowly increased to 1.1. The cell images taken before and after UM exposure indicate that the intensity reduction is more pronounced in those cells after exposure. Hence the results show the potential of UM as a non-invasive in vitro stimulation tool for facilitating targeted drug delivery and gene transfection as well as for studying cellular mechanotransduction.


Assuntos
Mecanotransdução Celular , Receptor ErbB-2/metabolismo , Ultrassom/métodos , Linhagem Celular Tumoral , Permeabilidade da Membrana Celular , Estudos de Viabilidade , Fluorescência , Humanos , Rodaminas , Transdutores , Óxido de Zinco
20.
J Biomed Opt ; 17(1): 015003, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22352647

RESUMO

We recently reported that a targeted, brightly fluorescent gallium corrole (HerGa) is highly effective for breast tumor detection and treatment. Unlike structurally similar porphryins, HerGa exhibits tumor-targeted toxicity without the need for photoexcitation. We have now examined whether photoexcitation further modulates HerGa toxicity, using multimode optical imaging of live cells, including two-photon excited fluorescence, differential interference contrast (DIC), spectral, and lifetime imaging. Using two-photon excited fluorescence imaging, we observed that light at specific wavelengths augments the HerGa-mediated mitochondrial membrane potential disruption of breast cancer cells in situ. In addition, DIC, spectral, and fluorescence lifetime imaging enabled us to both validate cell damage by HerGa photoexcitation and investigate HerGa internalization, thus allowing optimization of light dose and timing. Our demonstration of HerGa phototoxicity opens the way for development of new methods of cancer intervention using tumor-targeted corroles.


Assuntos
Corantes Fluorescentes/farmacologia , Gálio/farmacologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/efeitos da radiação , Porfirinas/farmacologia , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular , Corantes Fluorescentes/química , Corantes Fluorescentes/farmacocinética , Gálio/química , Gálio/farmacocinética , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Potencial da Membrana Mitocondrial/efeitos da radiação , Microscopia de Fluorescência/métodos , Mitocôndrias/patologia , Compostos Organometálicos/química , Processos Fotoquímicos/efeitos dos fármacos , Processos Fotoquímicos/efeitos da radiação , Porfirinas/química , Porfirinas/farmacocinética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA