Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 285
Filtrar
1.
J Cell Physiol ; 238(5): 1006-1019, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36870066

RESUMO

The engulfment adaptor phosphotyrosine-binding domain containing 1 (GULP1) is an adaptor protein involved in the engulfment of apoptotic cells via phagocytosis. Gulp1 was first found to promote the phagocytosis of apoptotic cells by macrophages, and its role in various tissues, including neurons and ovaries, has been well studied. However, the expression and function of GULP1 in bone tissue are poorly understood. Consequently, to determine whether GULP1 plays a role in the regulation of bone remodeling in vitro and in vivo, we generated Gulp1 knockout (KO) mice. Gulp1 was expressed in bone tissue, mainly in osteoblasts, while its expression is very low in osteoclasts. Microcomputed tomography and histomorphometry analysis in 8-week-old male Gulp1 KO mice revealed a high bone mass in comparison with male wild-type (WT) mice. This was a result of decreased osteoclast differentiation and function in vivo and in vitro as confirmed by a reduced actin ring and microtubule formation in osteoclasts. Gas chromatography-mass spectrometry analysis further showed that both 17ß-estradiol (E2) and 2-hydroxyestradiol levels, and the E2/testosterone metabolic ratio, reflecting aromatase activity, were also higher in the bone marrow of male Gulp1 KO mice than in male WT mice. Consistent with mass spectrometry analysis, aromatase enzymatic activity was significantly higher in the bone marrow of male Gulp1 KO mice. Altogether, our results suggest that GULP1 deficiency decreases the differentiation and function of osteoclasts themselves and increases sex steroid hormone-mediated inhibition of osteoclast differentiation and function, rather than affecting osteoblasts, resulting in a high bone mass in male mice. To the best of our knowledge, this is the first study to explore the direct and indirect roles of GULP1 in bone remodeling, providing new insights into its regulation.


Assuntos
Aromatase , Estradiol , Osteoclastos , Animais , Masculino , Camundongos , Aromatase/genética , Aromatase/metabolismo , Osso e Ossos , Diferenciação Celular , Camundongos Knockout , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Microtomografia por Raio-X , Estradiol/metabolismo
2.
Small ; 19(37): e2300527, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37226374

RESUMO

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


Assuntos
Vesículas Extracelulares , Neoplasias , Camundongos , Animais , Vesículas Extracelulares/metabolismo , Neoplasias/patologia , Citoplasma , Citosol , Imunoterapia/métodos
3.
Small ; 19(41): e2301431, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37282761

RESUMO

Manipulation and control of cell chemotaxis remain an underexplored territory despite vast potential in various fields, such as cytotherapeutics, sensors, and even cell robots. Herein is achieved the chemical control over chemotactic movement and direction of Jurkat T cells, as a representative model, by the construction of cell-in-catalytic-coat structures in single-cell nanoencapsulation. Armed with the catalytic power of glucose oxidase (GOx) in the artificial coat, the nanobiohybrid cytostructures, denoted as Jurkat[Lipo_GOx] , exhibit controllable, redirected chemotactic movement in response to d-glucose gradients, in the opposite direction to the positive-chemotaxis direction of naïve, uncoated Jurkat cells in the same gradients. The chemically endowed, reaction-based fugetaxis of Jurkat[Lipo_GOx] operates orthogonally and complementarily to the endogenous, binding/recognition-based chemotaxis that remains intact after the formation of a GOx coat. For instance, the chemotactic velocity of Jurkat[Lipo_GOx] can be adjusted by varying the combination of d-glucose and natural chemokines (CXCL12 and CCL19) in the gradient. This work offers an innovative chemical tool for bioaugmenting living cells at the single-cell level through the use of catalytic cell-in-coat structures.


Assuntos
Quimiotaxia , Glucose , Humanos , Células Jurkat , Glucose Oxidase , Catálise
4.
Int J Mol Sci ; 23(11)2022 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-35682801

RESUMO

The SARS-CoV-2 pandemic has created a global public crisis and heavily affected personal lives, healthcare systems, and global economies. Virus variants are continuously emerging, and, thus, the pandemic has been ongoing for over two years. Vaccines were rapidly developed based on the original SARS-CoV-2 (Wuhan-Hu-1) to build immunity against the coronavirus disease. However, they had a very low effect on the virus' variants due to their low cross-reactivity. In this study, a multivalent SARS-CoV-2 vaccine was developed using ferritin nanocages, which display the spike protein from the Wuhan-Hu-1, B.1.351, or B.1.429 SARS-CoV-2 on their surfaces. We show that the mixture of three SARS-CoV-2 spike-protein-displaying nanocages elicits CD4+ and CD8+ T cells and B-cell immunity successfully in vivo. Furthermore, they generate a more consistent antibody response against the B.1.351 and B.1.429 variants than a monovalent vaccine. This leads us to believe that the proposed ferritin-nanocage-based multivalent vaccine platform will provide strong protection against emerging SARS-CoV-2 variants of concern (VOCs).


Assuntos
COVID-19 , Vacinas Virais , Anticorpos Neutralizantes/genética , Linfócitos T CD8-Positivos , COVID-19/prevenção & controle , Vacinas contra COVID-19 , Ferritinas/genética , Humanos , Imunidade , Mutação , SARS-CoV-2 , Vacinas Combinadas
5.
Apoptosis ; 26(3-4): 209-218, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33655467

RESUMO

Apoptosis plays an essential role in the pathophysiologic processes of rheumatoid arthritis. A molecular probe that allows spatiotemporal observation of apoptosis in vitro, in vivo, and ex vivo concomitantly would be useful to monitoring or predicting pathophysiologic stages. In this study we investigated whether cyclic apoptosis-targeting peptide-1 (CApoPep-1) can be used as an apoptosis imaging probe in inflammatory arthritis. We tested the utility of CApoPep-1 for detecting apoptotic immune cells in vitro and ex vivo using flow cytometry and immunofluorescence. The feasibility of visualizing and quantifying apoptosis using this probe was evaluated in a murine collagen-induced arthritis (CIA) model, especially after treatment. CApoPep-1 peptide may successfully replace Annexin V for in vitro and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay for ex vivo in the measurement of apoptotic cells, thus function as a sensitive probe enough to be used clinically. In vivo imaging in CIA mice revealed that CApoPep-1 had 42.9 times higher fluorescence intensity than Annexin V for apoptosis quantification. Furthermore, it may be used as an imaging probe for early detection of apoptotic response in situ after treatment. The CApoPep-1 signal was mostly co-localized with the TUNEL signal (69.6% of TUNEL+ cells) in defined cell populations in joint tissues of CIA mice. These results demonstrate that CApoPep-1 is sufficiently sensitive to be used as an apoptosis imaging probe for multipurpose applications which could detect the same target across in vitro, in vivo, to ex vivo in inflammatory arthritis.


Assuntos
Artrite/diagnóstico por imagem , Diagnóstico por Imagem/métodos , Corantes Fluorescentes/química , Oligopeptídeos/química , Animais , Apoptose , Artrite Experimental/diagnóstico por imagem , Artrite Reumatoide/diagnóstico por imagem , Modelos Animais de Doenças , Marcação In Situ das Extremidades Cortadas/métodos , Camundongos
6.
Int J Mol Sci ; 22(23)2021 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-34884721

RESUMO

Cancer immunotherapy is fast rising as a prominent new pillar of cancer treatment, harnessing the immune system to fight against numerous types of cancer. Rho-kinase (ROCK) pathway is involved in diverse cellular activities, and is therefore the target of interest in various diseases at the cellular level including cancer. Indeed, ROCK is well-known for its involvement in the tumor cell and tumor microenvironment, especially in its ability to enhance tumor cell progression, migration, metastasis, and extracellular matrix remodeling. Importantly, ROCK is also considered to be a novel and effective modulator of immune cells, although further studies are needed. In this review article, we describe the various activities of ROCK and its potential to be utilized in cancer treatment, particularly in cancer immunotherapy, by shining a light on its activities in the immune system.


Assuntos
Imunoterapia , Terapia de Alvo Molecular , Neoplasias/enzimologia , Quinases Associadas a rho/metabolismo , Animais , Humanos , Neoplasias/imunologia , Neoplasias/terapia , Microambiente Tumoral
7.
Bioconjug Chem ; 31(5): 1562-1574, 2020 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-32369345

RESUMO

Exosomes are cellular components with promising uses in cancer diagnostics and therapeutics, and their imaging and tracking are essential to study their biological properties. Herein, we report on an in situ one-step fluorescence labeling strategy for exosomes via bioorthogonal click chemistry. First, exosome donor cancer cells were treated with tetraacetylated N-azidoacetyl-d-mannosamine (Ac4ManNAz) to generate unnatural azide groups (-N3) on their surface via metabolic glycoengineering. Then, the azide groups were labeled with near-infrared fluorescent dye-conjugated dibenzylcyclooctyne (DBCO-Cy5) via bioorthogonal click chemistry. After 2 days of incubation, the DBCO-Cy5-labeled exosomes (Cy5-Exo) were successfully secreted from the donor cancer cells and were isolated via classical ultracentrifugation, providing a high-yield of fluorescent dye-labeled exosomes. This in situ one-step bioorthogonal click chemistry offers improved labeling efficiency, biocompatibility, and imaging sensitivy compared to standard exosomes (ST-Exo), purified with classical ultracentrifugation or carbocyanine lipophilic dye (DiD)-labeled exosomes (DiD-Exo) in vitro. In particular, the Cy5-Exo were successfully taken up by A549 cells in a time-dependent manner, and they could escape from lysosome confinement, showing their possible use as a delivery carrier of therapeutic drugs or imaging agents. Finally, intraveneously injected Cy5-Exo were noninvasively tracked and imaged via near-infrared fluorescence (NIRF) imaging in tumor-bearing mice. This new fluorescence labeling strategy for natural exosomes may be useful to provide better understanding of their theranostic effects in many biomedical applications.


Assuntos
Exossomos/metabolismo , Corantes Fluorescentes/química , Corantes Fluorescentes/metabolismo , Imagem Óptica/métodos , Animais , Linhagem Celular Tumoral , Química Click , Cicloparafinas/química , Humanos , Camundongos
8.
Int J Mol Sci ; 21(19)2020 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-33008099

RESUMO

The placenta undergoes reconstruction at different times during fetal development to supply oxygen and nutrients required throughout pregnancy. To accommodate the rapid growth of the fetus, small spiral arteries undergo remodeling in the placenta. This remodeling includes apoptosis of endothelial cells that line spiral arteries, which are replaced by trophoblasts of fetal origin. Removal of dead cells is critical during this process. Stabilin-1 (Stab1) and stabilin-2 (Stab2) are important receptors expressed on scavenger cells that absorb and degrade apoptotic cells, and Stab1 is expressed in specific cells of the placenta. However, the role of Stab1 and Stab2 in placental development and maintenance remain unclear. In this study, we assessed Stab1 and Stab2 expression in the placenta and examined the reproductive capacity and placental development using a double-knockout mouse strain lacking both Stab1 and Stab2 (Stab1/2 dKO mice). Most pregnant Stab1/2 dKO female mice did not produce offspring and exhibited placental defects, including decidual hemorrhage and necrosis. Findings of this study offer the first description of the phenotypic characteristics of placentas and embryos of Stab1/2 dKO females during pregnancy, suggesting that Stab1 and Stab2 are involved in placental development and maintenance.


Assuntos
Moléculas de Adesão Celular Neuronais/genética , Placenta/metabolismo , Placentação/genética , Animais , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Feminino , Desenvolvimento Fetal/genética , Humanos , Camundongos , Camundongos Knockout , Oxigênio/metabolismo , Placenta/patologia , Gravidez , Reprodução/genética , Trofoblastos/metabolismo , Trofoblastos/patologia , Remodelação Vascular/genética
9.
Int J Mol Sci ; 21(4)2020 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-32102392

RESUMO

Exosomes are nanosized vesicles (30-140 nm) of endocytic origin that play important roles in regenerative medicine. They are derived from cell membranes during endocytic internalization and stabilize in biological fluids such as blood and synovia. Temporomandibular joint osteoarthritis (TMJ OA) is a degenerative disease, which, in addition to chronic pain, is characterized by progressive cartilage breakdown, condylar bone remodeling, and synovitis. However, traditional clinical treatments have limited symptom- and structure-modifying effects to restore damaged cartilage and other TMJ tissues. This is due to the limited self-healing capacity of condylar cartilage. Recently, stem-cell-derived exosomes have been studied as an alternative therapeutic approach to tissue repair and regeneration. It is known that trophic regulation of mesenchymal stem cells (MSCs) has anti-inflammatory and immunomodulatory effects under pathological conditions, and research on MSC-derived exosomes is rapidly accumulating. MSC-derived exosomes mimic the major therapeutic effects of MSCs. They affect the activity of immune effector cells and possess multilineage differentiation potential, including chondrogenic and osteogenic differentiation. Furthermore, exosomes are capable of regenerating cartilage or osseous compartments and restoring injured tissues and can treat dysfunction and pain caused by TMJ OA. In this review, we looked at the uniqueness of TMJ, the pathogenesis of TMJ OA, and the potential role of MSC-derived exosomes for TMJ cartilage and bone regeneration.


Assuntos
Exossomos/metabolismo , Células-Tronco Mesenquimais/metabolismo , Osteoartrite/metabolismo , Regeneração , Medicina Regenerativa/métodos , Articulação Temporomandibular/metabolismo , Animais , Humanos , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Osteoartrite/fisiopatologia , Osteogênese , Articulação Temporomandibular/patologia , Articulação Temporomandibular/fisiopatologia
10.
Gut ; 68(4): 693-707, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30415234

RESUMO

OBJECTIVE: Pancreatic cancer is associated with an abundant stromal reaction leading to immune escape and tumour growth. This massive stroma drives the immune escape in the tumour. We aimed to study the impact of ßig-h3 stromal protein in the modulation of the antitumoural immune response in pancreatic cancer. DESIGN: We performed studies with p48-Cre;KrasG12D, pdx1-Cre;KrasG12D;Ink4a/Arffl/fl, pdx1-Cre;KrasG12D; p53R172H mice and tumour tissues from patients with pancreatic ductal adenocarcinoma (PDA). Some transgenic mice were given injections of anti-ßig-h3, anti-CD8, anti-PD1 depleting antibodies. Tumour growth as well as modifications in the activation of local immune cells were analysed by flow cytometry, immunohistochemistry and immunofluorescence. Tissue stiffness was measured by atomic force microscopy. RESULTS: We identified ßig-h3 stromal-derived protein as a key actor of the immune paracrine interaction mechanism that drives pancreatic cancer. We found that ßig-h3 is highly produced by cancer-associated fibroblasts in the stroma of human and mouse. This protein acts directly on tumour-specific CD8+ T cells and F4/80 macrophages. Depleting ßig-h3 in vivo reduced tumour growth by enhancing the number of activated CD8+ T cell within the tumour and subsequent apoptotic tumour cells. Furthermore, we found that targeting ßig-h3 in established lesions released the tissue tension and functionally reprogrammed F4/80 macrophages in the tumour microenvironment. CONCLUSIONS: Our data indicate that targeting stromal extracellular matrix protein ßig-h3 improves the antitumoural response and consequently reduces tumour weight. Our findings present ßig-h3 as a novel immunological target in pancreatic cancer.


Assuntos
Adenocarcinoma/imunologia , Linfócitos T CD8-Positivos/imunologia , Carcinoma Ductal Pancreático/imunologia , Proteínas da Matriz Extracelular/imunologia , Neoplasias Pancreáticas/imunologia , Fator de Crescimento Transformador beta/imunologia , Microambiente Tumoral/imunologia , Animais , Fibroblastos/imunologia , Citometria de Fluxo , Imunofluorescência , Humanos , Imuno-Histoquímica , Macrófagos/imunologia , Camundongos , Camundongos Transgênicos , Microscopia de Força Atômica , Comunicação Parácrina/imunologia
11.
Calcif Tissue Int ; 105(2): 205-214, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31025051

RESUMO

Stabilin-1 is a transmembrane receptor that regulates molecule recycling and cell homeostasis by controlling the intracellular trafficking and participates in cell-cell adhesion and transmigration. Stabilin-1 expression is observed in various organs, including bones; however, its function and regulatory mechanisms in the bone remain unclear. In this study, we evaluated the physiological function of stabilin-1 in bone cells and tissue using a stabilin-1 knockout (Stab1 KO) mouse model. In wild-type (WT) mice, stabilin-1 was expressed in osteoblasts and osteoclasts, and its expression was maintained during osteoblast differentiation but significantly decreased after osteoclast differentiation. There was no difference in osteoblast differentiation and function, or the expression of osteoblast differentiation markers between mesenchymal stem cells isolated from Stab1 KO and WT mice. However, osteoclast differentiation marker levels demonstrated a non-significant increase and bone-resorbing activity was significantly increased in vitro in RANKL-induced osteoclasts from Stab1-deficient bone marrow macrophages (BMMs) compared with those of WT BMMs. Microcomputed tomography showed a negligible difference between WT and Stab1 KO mice in bone volume and trabecular thickness and number. Moreover, no in vivo functional defect in bone formation by osteoblasts was observed in the Stab1 KO mice. The osteoclast surface and number showed an increased tendency in Stab1 KO mice compared to WT mice in vivo, but this difference was not statistically significant. Overall, these results indicate that Stab1 does not play an essential role in in vivo bone development and bone cell function, but it does affect in vitro osteoclast maturation and function for bone resorption.


Assuntos
Moléculas de Adesão Celular Neuronais/genética , Moléculas de Adesão Celular Neuronais/metabolismo , Osteoclastos/citologia , Animais , Células da Medula Óssea/citologia , Reabsorção Óssea , Osso e Ossos , Adesão Celular , Diferenciação Celular , Linhagem Celular , Movimento Celular , Feminino , Genótipo , Macrófagos/citologia , Masculino , Camundongos , Camundongos Knockout , Osteoblastos/citologia , Osteócitos/citologia , Osteogênese , Microtomografia por Raio-X
12.
Nanomedicine ; 14(3): 633-642, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29309907

RESUMO

The use of thrombolytic therapies is limited by an increased risk of systemic hemorrhage due to lysis of hemostatic clots. We sought to develop a plasmin-based thrombolytic nanocage that efficiently dissolves the clot without causing systemic fibrinolysis or disrupting hemostatic clots. Here, we generated a double chambered short-length ferritin (sFt) construct that has an N-terminal region fused to multivalent clot targeting peptides (CLT: CNAGESSKNC) and a C-terminal end fused to a microplasmin (µPn); CLT recognizes fibrin-fibronectin complexes in clots, µPn efficiently dissolves clots, and the assembly of double chambered sFt (CLT-sFt-µPn) into nanocage structure protects the activated-µPn from its circulating inhibitors. Importantly, activated CLT-sFt-µPn thrombolytic nanocage showed a prolonged circulatory life over activated-µPn and efficiently lysed the preexisting clots in both arterial and venous thromboses models. Thus, CLT-sFt-µPn thrombolytic nanocage platform represents the prototype of a targeted clot-busting agent with high efficacy and safety over existing thrombolytic therapies.


Assuntos
Trombose Coronária/prevenção & controle , Ferritinas/química , Fibrinolisina/química , Fibrinolíticos/administração & dosagem , Nanopartículas/administração & dosagem , Fragmentos de Peptídeos/química , Terapia Trombolítica/métodos , Trombose Venosa/prevenção & controle , Animais , Trombose Coronária/patologia , Modelos Animais de Doenças , Fibrinolíticos/química , Masculino , Camundongos , Camundongos Endogâmicos ICR , Nanopartículas/química , Ratos , Ratos Sprague-Dawley , Trombose Venosa/patologia
13.
J Cell Biochem ; 118(7): 1827-1838, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28059467

RESUMO

Numerous studies have implied that mutY DNA glycosylase (MYH) is involved in the repair of post-replicative mispairs and plays a critical role in the base excision repair pathway. Recent in vitro studies have shown that MYH interacts with tumor necrosis factor receptor type 1-associated death domain (TRADD), a key effector protein of tumor necrosis factor receptor-1 (TNFR1) signaling. The association between MYH and TRADD is reversed during tumor necrosis factor alpha (TNF-α)- and camptothecin (CPT)-induced apoptosis, and enhanced during TNF-α-induced survival. After investigating the role of MYH interacts with various proteins following TNF-α stimulation, here, we focus on MYH and TRADD interaction functions in necroptosis and its effects to related proteins. We report that the level of the MYH and TRADD complex was also reduced during necroptosis induced by TNF-α and zVAD-fmk. In particular, we also found that MYH is a biologically important necrosis suppressor. Under combined TNF-α and zVAD-fmk treatment, MYH-deficient cells were induced to enter the necroptosis pathway but primary mouse embryonic fibroblasts (MEFs) were not. Necroptosis in the absence of MYH proceeds via the inactivation of caspase-8, followed by an increase in the formation of the kinase receptor- interacting protein 1 (RIP1)-RIP3 complex. Our results suggested that MYH, which interacts with TRADD, inhibits TNF-α necroptotic signaling. Therefore, MYH inactivation is essential for necroptosis via the downregulation of caspase-8. J. Cell. Biochem. 118: 1827-1838, 2017. © 2017 Wiley Periodicals, Inc.


Assuntos
Apoptose/efeitos dos fármacos , DNA Glicosilases/metabolismo , Necrose/induzido quimicamente , Fator de Necrose Tumoral alfa/farmacologia , Clorometilcetonas de Aminoácidos/farmacologia , Animais , Western Blotting , Camptotecina/farmacologia , Caspase 8/metabolismo , Linhagem Celular , Células Cultivadas , DNA Glicosilases/genética , Imunofluorescência , Imunoprecipitação , Camundongos , Ligação Proteica , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Transdução de Sinais , Proteína de Domínio de Morte Associada a Receptor de TNF/metabolismo
14.
Int J Cancer ; 141(9): 1912-1920, 2017 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-28635011

RESUMO

Chemotherapy have commonly been used in maximum tolerated dose to completely eradicate the cancer. However, such treatments often failed due to the complex and dynamic nature of cancer. Therefore, it has been suggested that cancer should be treated as a chronic disease, controlling its growth by providing continuous therapeutic pressure for long-term. Such an approach, however, requires a therapy that is non-toxic and orally available with sufficient potency. Herein, we propose a radiotherapy-assisted orally available metronomic apoptosis-targeted chemotherapy, which delivers doxorubicin continuously to the irradiated tumor with high selectivity while causing minimal toxicities to the normal tissues. DEVD-S-DOX/DCK complex is the anticancer prodrug for our strategy that could selectively release doxorubicin in the irradiated tumor tissue with sufficient oral bioavailability. The prodrug was completely inactive by itself, but displayed potent anticancer activity when coupled with radiotherapy. Consequently, the daily oral administration of DEVD-S-DOX/DCK in combination with the low-dose radiotherapy effectively suppressed the growth of tumor in vivo with no significant systemic toxicities despite that the accumulated dose of doxorubicin exceeded 150 mg/kg. Therefore, the our novel therapy using DEVD-S-DOX/DCK complex is considered as an outstanding treatment option for treating cancer for long-term attributed to its oral availability and low-toxicity profile as well as the potent anticancer effect.


Assuntos
Doxorrubicina/administração & dosagem , Neoplasias/tratamento farmacológico , Neoplasias/radioterapia , Pró-Fármacos/administração & dosagem , Administração Oral , Animais , Apoptose/efeitos dos fármacos , Células CACO-2 , Terapia Combinada , Doxorrubicina/química , Sistemas de Liberação de Medicamentos , Humanos , Dose Máxima Tolerável , Camundongos , Neoplasias/patologia , Pró-Fármacos/química , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Biomacromolecules ; 17(1): 12-9, 2016 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-26646195

RESUMO

Ferritin cage nanoparticles are promising platforms for targeted delivery of imaging and therapeutic agents. One of the main advantages of cage nanoparticles is the ability to display multiple functionalities through genetic modification so as to achieve desired therapeutic or diagnostic functions. Ferritin complexes formed from short ferritin (sFt) lacking the fifth helix can accommodate dual peptide and protein functionalities on N- and C-terminal sites in sFt monomers. The resulting double-chambered NanoCage (DCNC) offers the potential of dual activities; these activities are augmented by the avidity of the ligands, which do not impede each other's function. Here we demonstrated proof-of-concept of DCNCs, loading the tumor-targeting proapoptotic peptide CGKRK(KLAKLAK)2 onto the N-terminal chamber and green fluorescent protein (GFP) onto the C-terminal chamber. The resulting KLAK-sFt-GFP DCNCs were internalized into the human breast adenocarcinoma cell line MDA-MB-231 and induced apoptosis. These findings suggest that DCNCs containing various combinations of peptides and proteins could be applied as therapeutics in different diseases.


Assuntos
Ferritinas/química , Proteínas de Fluorescência Verde/química , Peptídeos/química , Animais , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos/métodos , Feminino , Humanos , Ligantes , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Nanopartículas/química , Nanotecnologia/métodos
16.
Biomacromolecules ; 17(3): 1150-9, 2016 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-26899206

RESUMO

Ferritin cage nanoparticles are promising platforms for targeted delivery of imaging and therapeutic agents because their cage structure can accommodate small molecules and their surfaces can be decorated with multiple functionalities. However, selective targeting is still a challenge for translating ferritin-based nanomedicines into the clinic, especially for heterogeneous diseases such as cancer. Targeting peptides can be genetically fused onto the surface of a ferritin cage, forming peptide bunches on nanocages (PBNCs) that offer synergistic increases in binding avidity. Here, we utilized two sites of the ferritin monomer, the N-terminus and the loop between the fourth and fifth helices, which are exposed on the surface of the assembled 24-subunit ferritin cage, to ligate one or two types of peptides to achieve "super affinity" and bispecificity, respectively. PBNCs formed by ligation of the IL-4 receptor-targeting peptide, AP1, to both sites (48AP1-PBNCs) tethered IL-4R, expressing tumor cells with greater affinity than did PBNCs with AP1 ligated to a single site (24AP1-PBNCs). Moreover, bispecific PBNCs containing 24 RGD peptides and 24 AP1 peptides (24RGD/24AP1-PBNCs) were capable of independently targeting cells expressing the corresponding receptors. Bispecific and superaffinity PBNCs could be useful for efficient targeting of ferritin-based therapeutic/diagnostic agents in a clinical setting.


Assuntos
Ferritinas/química , Nanopartículas Metálicas/química , Oligopeptídeos/química , Linhagem Celular Tumoral , Humanos , Ligantes , Oligopeptídeos/metabolismo , Ligação Proteica , Receptores de Interleucina-4/metabolismo
17.
Apoptosis ; 20(1): 110-21, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25430587

RESUMO

Apoptosis has a role in many medical disorders and treatments; hence, its non-invasive evaluation is one of the most riveting research topics. Currently annexin V is used as gold standard for imaging apoptosis. However, several drawbacks, including high background, slow body clearance, make it a suboptimum marker for apoptosis imaging. In this study, we radiolabeled the recently identified histone H1 targeting peptide (ApoPep-1) and evaluated its potential as a new apoptosis imaging agent in various animal models. ApoPep-1 (CQRPPR) was synthesized, and an extra tyrosine residue was added to its N-terminal end for radiolabeling. This peptide was radiolabeled with (124)I and (131)I and was tested for its serum stability. Surgery- and drug-induced apoptotic rat models were prepared for apoptosis evaluation, and PET imaging was performed. Doxorubicin was used for xenograft tumor treatment in mice, and the induced apoptosis was studied. Tumor metabolism and proliferation were assessed by [(18)F]FDG and [(18)F]FLT PET imaging and compared with ApoPep-1 after doxorubicin treatment. The peptide was radiolabeled at high purity, and it showed reasonably good stability in serum. Cell death was easily imaged by radiolabeled ApoPep-1 in an ischemia surgery model. And, liver apoptosis was more clearly identified by ApoPep-1 rather than [(124)I]annexin V in cycloheximide-treated models. Three doxorubicin doses inhibited tumor growth, which was evaluated by 30-40% decreases of [(18)F]FDG and [(18)F]FLT PET uptake in the tumor area. However, ApoPep-1 demonstrated more than 200% increase in tumor uptake after chemotherapy, while annexin V did not show any meaningful uptake in the tumor compared with the background. Biodistribution data were also in good agreement with the microPET imaging results. All of the experimental data clearly demonstrated high potential of the radiolabeled ApoPep-1 for in vivo apoptosis imaging.


Assuntos
Apoptose , Radioisótopos do Iodo , Neoplasias Pulmonares/patologia , Imagem Molecular , Animais , Antibióticos Antineoplásicos/uso terapêutico , Biomarcadores/metabolismo , Linhagem Celular Tumoral , Modelos Animais de Doenças , Doxorrubicina/uso terapêutico , Xenoenxertos , Histonas/química , Histonas/metabolismo , Humanos , Marcação por Isótopo , Neoplasias Pulmonares/tratamento farmacológico , Camundongos Endogâmicos BALB C , Camundongos Nus , Peptídeos/química , Peptídeos/metabolismo , Ratos Sprague-Dawley
18.
Calcif Tissue Int ; 96(1): 56-64, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25450762

RESUMO

Transforming growth factor-ß-induced gene product-h3 (TGFBI/BIGH3) is an extracellular matrix protein expressed in a wide variety of tissues. TGFBI binds to type I, II, and IV collagens, as well as to biglycan and decorin and plays important roles in cell-to-cell, cell-to-collagen, and cell-to-matrix interactions. Furthermore, TGFBI is involved in cell growth and migration, tumorigenesis, wound healing, and apoptosis. To investigate whether TGFBI is involved in the maintenance of skeletal tissues, Tgfbi knockout mice were generated by crossing male and female Tgfbi heterozygous mice. Skeletal preparation showed that the skeletal size in Tgfbi knockout mice was smaller than in wild-type and heterozygous mice. However, chondrocytic cell alignment in the growth plates, bone mineral density, and bone forming rates were similar in Tgfbi knockout, wild-type, and heterozygous mice. Alterations in skeletal tissue arrangements in Tgfbi knockout mice were estimated from safranin O staining, trichrome staining, and immunohistochemistry for type II and X collagen, and matrix metalloproteinase 13 (MMP13). Cartilage matrix degradation was observed in the articular cartilage of Tgfbi knockout mice. Although the detection of type II collagen in the articular cartilage was lower in Tgfbi knockout mice than wild-type mice, the detection of MMP13 was markedly higher, indicating that Tgfbi deficiency is associated with the degradation of cartilage matrix. These results suggest that TGFBI plays an important role in maintaining skeletal tissues and the cartilage matrix in mice.


Assuntos
Densidade Óssea/genética , Matriz Óssea/metabolismo , Cartilagem Articular/patologia , Proteínas da Matriz Extracelular/genética , Fator de Crescimento Transformador beta/genética , Animais , Densidade Óssea/fisiologia , Matriz Óssea/patologia , Cartilagem Articular/metabolismo , Colágeno Tipo II/metabolismo , Metaloproteinase 13 da Matriz/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout
19.
Am J Respir Crit Care Med ; 189(7): 779-86, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24506343

RESUMO

RATIONALE: Sepsis is a systemic inflammatory condition resulting from bacterial infections; it has a high mortality rate and limited therapeutic options. Despite extensive research into the mechanisms driving bacterial sepsis, the target molecules controlling vascular leakage are still largely unknown. Transforming growth factor ß-induced protein (TGFBIp) is an extracellular matrix protein expressed in several cell types, which is known to interact with integrins. OBJECTIVES: The aim of this study was to determine the roles of TGFBIp in vascular proinflammatory responses, and the mechanisms of action driving these responses. METHODS: Circulating levels of TGFBIp were measured in patients admitted to the hospital with sepsis, severe sepsis, and septic shock and in cecal ligation and puncture (CLP)-induced septic mice. Effects of TGFBIp knockout on CLP-induced septic mortality and effects of TGFBIp on multiple vascular proinflammatory responses were determined. MEASUREMENTS AND MAIN RESULTS: Circulating levels of TGFBIp were significantly elevated compared with healthy controls, and were strongly correlated with disease severity. High blood TGFBIp levels were also observed in CLP-induced septic mice. The absence of the TGFBIp gene in mice attenuated CLP-induced sepsis. TGFBIp enhanced vascular proinflammatory responses including vascular permeability, adhesion and migration of leukocytes, and disruption of adherence junctions through interacting with integrin αvß5. CONCLUSIONS: Collectively, our findings demonstrate that the TGFBIp-αvß5 axis can elicit severe inflammatory responses, suggesting it to be a potential target for development of diagnostics and therapeutics for sepsis.


Assuntos
Endotélio Vascular/fisiopatologia , Proteínas da Matriz Extracelular/sangue , Sepse/fisiopatologia , Fator de Crescimento Transformador beta/sangue , Animais , Biomarcadores/sangue , Estudos de Casos e Controles , Ceco/cirurgia , Endotélio Vascular/metabolismo , Humanos , Leucócitos/fisiologia , Ligadura , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Permeabilidade , Receptores de Vitronectina/sangue , Sepse/sangue , Sepse/etiologia , Sepse/mortalidade , Índice de Gravidade de Doença
20.
Biochim Biophys Acta ; 1833(10): 2378-88, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23792174

RESUMO

TGFBI, a transforming growth factor ß-induced extracellular matrix protein, circulates at a level of ~300ng/ml in humans and modulates several integrin-mediated cellular functions. The protein contains an N-terminal EMI domain, four consecutive FAS1 domains, and the RGD motif. Each FAS1 domain and the RGD motif have been known to interact with avb3 integrin. Here, we found that the binding affinity (Kd) of TGFBI for αvß3 integrin was approximately 3.8×10(-8)M, a value ~2300-fold higher than that of a single FAS1 domain, and demonstrated that this greater affinity was due to the cooperative action of the four FAS1 domains and the RGD motif. Moreover, TGFBI exhibited more potent anti-angiogenic and anti-tumorigenic activities, even at a 100-fold lower molar dose than the reported effective dose of the FAS1 domain. Finally, our data showed that TGFBI specifically targeted the tumor vasculature and accumulated at the tumor site. Collectively, our results support the theory that TGFBI acts as a potent endogenous anti-tumor and anti-angiogenic molecule by targeting αvß3 integrin, and highlights the importance of physiological circulating TGFBI levels in inhibiting tumor growth.


Assuntos
Carcinoma Pulmonar de Lewis/prevenção & controle , Proliferação de Células , Proteínas da Matriz Extracelular/metabolismo , Integrina alfaVbeta3/metabolismo , Melanoma Experimental/prevenção & controle , Neovascularização Patológica/prevenção & controle , Oligopeptídeos/química , Fator de Crescimento Transformador beta/metabolismo , Receptor fas/química , Animais , Anticorpos Monoclonais/imunologia , Formação de Anticorpos , Western Blotting , Carcinoma Pulmonar de Lewis/irrigação sanguínea , Carcinoma Pulmonar de Lewis/patologia , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Proteínas da Matriz Extracelular/imunologia , Humanos , Técnicas Imunoenzimáticas , Masculino , Melanoma Experimental/irrigação sanguínea , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Oligopeptídeos/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/imunologia , Células Tumorais Cultivadas , Receptor fas/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa