Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Stem Cells ; 40(9): 818-830, 2022 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-35728620

RESUMO

Nerve growth factor (NGF) is the best-characterized neurotrophin and is primarily recognized for its key role in the embryonic development of the nervous system and neuronal cell survival/differentiation. Recently, unexpected actions of NGF in bone regeneration have emerged as NGF is able to enhance the osteogenic differentiation of mesenchymal stem cells. However, little is known regarding how NGF signaling regulates osteogenic differentiation through epigenetic mechanisms. In this study, using human dental mesenchymal stem cells (DMSCs), we demonstrated that NGF mediates osteogenic differentiation through p75NTR, a low-affinity NGF receptor. P75NTR-mediated NGF signaling activates the JNK cascade and the expression of KDM4B, an activating histone demethylase, by removing repressive H3K9me3 epigenetic marks. Mechanistically, NGF-activated c-Jun binds to the KDM4B promoter region and directly upregulates KDM4B expression. Subsequently, KDM4B directly and epigenetically activates DLX5, a master osteogenic gene, by demethylating H3K9me3 marks. Furthermore, we revealed that KDM4B and c-Jun from the JNK signaling pathway work in concert to regulate NGF-mediated osteogenic differentiation through simultaneous recruitment to the promoter region of DLX5. We identified KDM4B as a key epigenetic regulator during the NGF-mediated osteogenesis both in vitro and in vivo using the calvarial defect regeneration mouse model. In conclusion, our study thoroughly elucidated the molecular and epigenetic mechanisms during NGF-mediated osteogenesis.


Assuntos
Células-Tronco Mesenquimais , Osteogênese , Animais , Diferenciação Celular/genética , Epigênese Genética , Histona Desmetilases/metabolismo , Humanos , Histona Desmetilases com o Domínio Jumonji/genética , Histona Desmetilases com o Domínio Jumonji/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos , Fator de Crescimento Neural/genética , Fator de Crescimento Neural/metabolismo , Osteogênese/genética , Receptor de Fator de Crescimento Neural/genética , Receptor de Fator de Crescimento Neural/metabolismo
2.
Bone Res ; 10(1): 3, 2022 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-34992221

RESUMO

Osteoporosis is a highly prevalent public health burden associated with an increased risk of bone fracture, particularly in aging women. Estrogen, an important medicinal component for the preventative and therapeutic treatment of postmenopausal osteoporosis, induces osteogenesis by activating the estrogen receptor signaling pathway and upregulating the expression of osteogenic genes, such as bone morphogenetic proteins (BMPs). The epigenetic regulation of estrogen-mediated osteogenesis, however, is still unclear. In this report, we found that estrogen significantly induced the expression of lysine-specific demethylase 6B (KDM6B) and that KDM6B depletion by shRNAs led to a significant reduction in the osteogenic potential of DMSCs. Mechanistically, upon estrogen stimulation, estrogen receptor-α (ERα) was recruited to the KDM6B promoter, directly enhancing KDM6B expression. Subsequently, KDM6B was recruited to the BMP2 and HOXC6 promoters, resulting in the removal of H3K27me3 marks and activating the transcription of BMP2 and HOXC6, the master genes of osteogenic differentiation. Furthermore, we found that estrogen enhanced DMSC osteogenesis during calvarial bone regeneration and that estrogen's pro-osteogenic effect was dependent on KDM6B in vivo. Taken together, our results demonstrate the vital role of the ERα/KDM6B regulatory axis in the epigenetic regulation of the estrogen-dependent osteogenic response.

3.
Sci Rep ; 10(1): 6383, 2020 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-32286430

RESUMO

Periodontitis is a local and systemic inflammatory condition and a risk factor of atherosclerosis, but no studies investigated the effect of a statin on atherogenesis affected by severe periodontitis. In this study, we investigated the effect of rosuvastatin (RSV) on atherogenesis in Apolipoprotein E-deficient mice receiving silk ligature placement around the maxillary second molars. Mice with the ligature placement developed severe periodontitis and vascular inflammation. RSV significantly inhibited the development of periodontitis and vascular inflammation and remarkably blocked the increased lipid deposition and the atherogenic gene expression in the arterial wall and aortic sinus induced by severe periodontitis. To understand the mechanistic effect of RSV on periodontitis-associated atherogenesis, we investigated the in vitro effect of RSV on various effect of TNF-α, a major proinflammatory cytokine for periodontitis and atherogenesis. We found that RSV notably inhibited the TNF-α-induced osteoclast formation, endothelial cell phenotypic changes, foam cell formation, and the expression of CD47 and other oncogenes in arterial smooth muscle cells. Taken together, our study indicates that RSV prevents the exacerbation of atherosclerosis induced periodontitis by inhibiting local, systemic and vascular inflammation, as well as the expression of CD47 from arterial smooth muscle cells in mice.


Assuntos
Aterosclerose/tratamento farmacológico , Inflamação/tratamento farmacológico , Periodontite/complicações , Rosuvastatina Cálcica/uso terapêutico , Animais , Aterosclerose/etiologia , Linhagem Celular , Citocinas/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Masculino , Camundongos , Camundongos Knockout para ApoE , Osteoclastos/efeitos dos fármacos , Osteoclastos/patologia , Seio Aórtico/efeitos dos fármacos
4.
J Biomed Mater Res A ; 106(2): 531-542, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28975732

RESUMO

Bioactive agents, including proteins and peptides, can be loaded into hydrogels to improve bone regenerative capacity with their controlled release. However, the current loading method has focused on physical mixing, which has limited release control. Therefore, alternative conjugation of bioactive agents with hydrogels is highly recommended. Direct chemical conjugation of synthetic peptides containing a functional moiety with a hydrogel would be ideal. Here, we synthesized a bioactive calcium accumulating peptide (CAP) containing a collagen binding motif, which can induce osteogenic differentiation. A tyrosine residue in CAP was used to directly chemically conjugate the peptide with a gelatin-based enzymatically crosslinked hydroxyphenyl propionic acid hydrogel under H2 O2 /Horse radish peroxidase conditions. To test the acceleration of bone formation, human periodontal ligament stem cells (PDLSCs) were loaded into a chemically conjugated CAP hydrogel. The CAP hydrogel induced bone mineralization around the PDLSCs and increased osteogenic marker expressions in vitro. It also recovered a bone layer in a calvarial defect 4 weeks postimplantation. In summary, an injectable CAP hydrogel scaffold system was developed as a potentially useful engineered microenvironment to enhance bone restoration, and it could be utilized as a vehicle for bioactive delivery of stem cells in tissue regenerative therapy. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 531-542, 2018.


Assuntos
Regeneração Óssea/efeitos dos fármacos , Cálcio/farmacologia , Gelatina/farmacologia , Hidrogéis/farmacologia , Peptídeos/farmacologia , Sequência de Aminoácidos , Animais , Proteína Morfogenética Óssea 2/farmacologia , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Humanos , Osteocalcina/metabolismo , Osteogênese/efeitos dos fármacos , Osteopontina/química , Peptídeos/síntese química , Peptídeos/química , Ligamento Periodontal/citologia , Ratos Sprague-Dawley , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Células-Tronco/ultraestrutura
5.
Biochem Biophys Res Commun ; 491(3): 827-833, 2017 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-28554844

RESUMO

Cancer stem cells (CSCs) are a subpopulation of cancer cells and have been known to create cancer reoccurrence during cancer therapy due to their stem cell-like characteristics. However, exact target to control the CSC has not been fully established. Here, we enriched CD44High population of MDA-MB-231 cells by CD44 antibody as a CSC marker. By Phospho Antibody Array, CD44High population of MDA-MB-231 cells reveals Feline sarcoma-related tyrosine kinase (FER) protein was highly activated. When FER siRNA and low molecular weight protamine (LMWP) as cell penetrating peptides are applied to this population, cancer migration and colony forming ability are inhibited. Moreover, silencing FER using FER siRNA and LMWP conjugates enhances anti-metastasis related factors including E-cadherin, p75 and p63. Taken together, FER is a new marker for targeting breast CSCs and peptide-mediated siRNA method could be an effective and safe way of delivery and be a new therapeutic strategy for targeting breast cancer.


Assuntos
Apoptose/efeitos dos fármacos , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/fisiologia , Peptídeos/administração & dosagem , Proteínas Tirosina Quinases/genética , RNA Interferente Pequeno/uso terapêutico , Apoptose/genética , Linhagem Celular Tumoral , Inativação Gênica , Marcação de Genes/métodos , Terapia Genética/métodos , Humanos , Terapia de Alvo Molecular/métodos , Peptídeos/farmacocinética , Proteínas Tirosina Quinases/antagonistas & inibidores , RNA Interferente Pequeno/genética , Resultado do Tratamento
6.
Int J Nanomedicine ; 11: 4643-4656, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27695323

RESUMO

Angiogenesis plays a critical role in the growth and metastasis of cancer, and growth factors released from cancer promote blood-vessel formation in the tumor microenvironment. The angiogenesis is accelerated via interactions of growth factors with the high-affinity receptors on cancer cells. In particular, heparan sulfate proteoglycans (HSPGs) on the surface of cancer cells have been shown to be important in many aspects of determining a tumor's phenotype and development. Specifically, the regulation of the interactions between HSPGs and growth factors results in changes in tumor progression. A peptide with heparin-binding (HBP) activity has been developed and synthesized to inhibit tumor growth via the prevention of angiogenesis. We hypothesized that HBP could inhibit the interaction of growth factors and HSPGs on the surface of cancer cells, decrease paracrine signaling in endothelial cells (ECs), and finally decrease angiogenesis in the tumor microenvironment. In this study, we found that HBP had antiangiogenic effects in vitro and in vivo. The conditioned media obtained from a breast cancer cell line treated with HBP were used to culture human umbilical vein ECs (HUVECs) to evaluate the antiangiogenic effect of HBP on ECs. HBP effectively inhibited the migration, invasion, and tube formation of HUVECs in vitro. In addition, the expressions of angiogenesis-mediating factors, including ERK, FAK, and Akt, were considerably decreased. HBP also decreased the levels of invasive factors, including MMP2 and MMP9, secreted by the HUVECs. We demonstrated significant suppression of tumor growth in a breast cancer xenograft model and enhanced distribution of HBP at the site of tumors. Taken together, our results show that HBP has antiangiogenic effects on ECs, and suggest that it may serve as a potential antitumor agent through control of the tumor microenvironment.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Proteína Morfogenética Óssea 4/uso terapêutico , Neoplasias/irrigação sanguínea , Neoplasias/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Peptídeos/uso terapêutico , Sequência de Aminoácidos , Inibidores da Angiogênese/farmacologia , Animais , Proteína Morfogenética Óssea 4/química , Proteína Morfogenética Óssea 4/farmacologia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Feminino , Heparina/metabolismo , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Metaloproteinase 2 da Matriz/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias/metabolismo , Peptídeos/química , Peptídeos/farmacologia , Transdução de Sinais/efeitos dos fármacos , Sus scrofa , Distribuição Tecidual/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Int J Nanomedicine ; 10: 5423-34, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26347021

RESUMO

Human beta-defensins (hBDs) are crucial factors of intrinsic immunity that function in the immunologic response to a variety of invading enveloped viruses, bacteria, and fungi. hBDs can cause membrane depolarization and cell lysis due to their highly cationic nature. These molecules participate in antimicrobial defenses and the control of adaptive and innate immunity in every mammalian species and are produced by various cell types. The C-terminal 15-mer peptide within hBD3, designated as hBD3-3, was selected for study due to its cell- and skin-penetrating activity, which can induce anti-inflammatory activity in lipopolysaccharide-treated RAW 264.7 macrophages. hBD3-3 penetrated both the outer membrane of the cells and mouse skin within a short treatment period. Two other peptide fragments showed poorer penetration activity compared to hBD3-3. hBD3-3 inhibited the lipopolysaccharide-induced production of inducible nitric oxide synthase, nitric oxide, and secretory cytokines, such as interleukin-6 and tumor necrosis factor in a concentration-dependent manner. Moreover, hBD3-3 reduced the interstitial infiltration of polymorphonuclear leukocytes in a lung inflammation model. Further investigation also revealed that hBD3-3 downregulated nuclear factor kappa B-dependent inflammation by directly suppressing the degradation of phosphorylated-IκBα and by downregulating active nuclear factor kappa B p65. Our findings indicate that hBD3-3 may be conjugated with drugs of interest to ensure their proper translocation to sites, such as the cytoplasm or nucleus, as hBD3-3 has the ability to be used as a carrier, and suggest a potential approach to effectively treat inflammatory diseases.


Assuntos
Anti-Inflamatórios/química , Peptídeos Penetradores de Células/química , NF-kappa B/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , beta-Defensinas/metabolismo , Animais , Sobrevivência Celular , Citocinas/metabolismo , Humanos , Imunidade Inata/efeitos dos fármacos , Inflamação/metabolismo , Interleucina-6/metabolismo , Leucócitos/citologia , Lipopolissacarídeos/química , Macrófagos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Nus , Microscopia de Fluorescência , Óxido Nítrico/metabolismo , Fosforilação , Estrutura Terciária de Proteína , Ratos , Ratos Wistar , Fator de Necrose Tumoral alfa/metabolismo
8.
Biomaterials ; 35(37): 9747-9754, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25205451

RESUMO

Mineralization in mammalian cells is accomplished by concerted regulation of protein-based extracellular matrix (ECM) components, such as non-collagenous proteins and collagen fibrils. In this study, we investigated the ability of a collagen-binding motif (CBM) peptide derived from osteopontin to selectively affect osteogenic or adipogenic differentiation in vitro and in vivo. In particular, increased osteogenic differentiation and decreased adipogenic differentiation were observed in human mesenchymal stem cells (hMSCs). Osteocalcin (OCN) protein expression in MC3T3-E1 cells without osteogenic inducers was then investigated following treatment with the CBM peptide. In ovariectomized (OVX) mice, estrogen deficiency induced osteoporosis and increased fat tissue deposition. However, after the CBM peptide or estradiol was injected into the OVX mice for 2 months, the increased serum OCN concentration and alkaline phosphate (ALP) activity were decreased in the estradiol-treated group (OVX-E) and the high-concentration CBM peptide-treated group (OVX-HP). Significant bone loss was also observed in the ovariectomized mice (OVX-PBS). In particular, the bone volume per total volume (BV/TV) and bone mineral density (BMD) were significantly decreased in the OVX mice; however, both of these markers were restored in the OVX-HP group, which also had significantly well-developed bone structure and bone formation. In contrast to the bone structural change, adipose tissue was increased in the OVX-PBS. However, a significant decrease in total fat and subcutaneous fat was observed in the low-concentration CBM peptide-treated group (OVX-LP) and the estradiol-treated group (OVX-E). Taken together, these results suggest that the CBM peptide could be an effective therapeutic agent for osteoporosis due to its selective stimulation of osteogenic differentiation, rather than adipogenesis.


Assuntos
Colágeno/metabolismo , Osteogênese/efeitos dos fármacos , Osteopontina/química , Osteoporose/tratamento farmacológico , Peptídeos/química , Peptídeos/uso terapêutico , Células 3T3 , Adipogenia/efeitos dos fármacos , Animais , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Calcificação Fisiológica/efeitos dos fármacos , Linhagem Celular , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Osteoporose/metabolismo , Osteoporose/patologia , Peptídeos/farmacologia
9.
Biomaterials ; 35(24): 6287-98, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24831974

RESUMO

Targeting tissues/cells using probing materials to detect diseases such as cancer and inflammatory disease has been attempted with some success. Most of the molecular targets used in diagnosis and therapy were identified through the discovery of intracellular signaling pathways. Among intracellular signaling processes, the ubiquitination of proteins, and thereby their proteasomal degradation, is important because it plays a role in most diseases involving alterations to a component of the ubiquitination system, particularly E3 ligases, which have selective target-binding affinity and are key to the success of regulating the disorder. The regulation and monitoring of E3 ligases can be achieved using peptides containing protein-protein binding motifs. We generated a human protein-derived peptide that could target Smurf1, a member of the E3 ligase family, by competitively binding to osteo-Smads. To effectively deliver it into cells, the peptide was further modified with a cell-penetrating peptide. The peptide contains two fluorescent dyes: fluorescein isothiocyanate (FITC; absorbance/emission wavelengths: 495/519 nm) as a fluorophore and black hole quencher-1 (BHQ-1) as a fluorescence quencher. When the target Smurf1 combined with complementary sequences in the peptide probe, the distance between the fluorophore and BHQ-1 increased via a conformational change, resulting in the recovery of the fluorescence signal. Simultaneously, the degradation of Smad1/5/8 was blocked by the binding of the peptide probe to Smurf1, leading to the potentiation of the osteogenic pathway, which was reflected by an increase in the expression of osteoinductive genes, such as alkaline phosphatase and osteocalcin. Possible future applications of the peptide probe include its integration into imaging tools for the diagnosis of Smurf1-overexpressing diseases.


Assuntos
Permeabilidade da Membrana Celular/efeitos dos fármacos , Peptídeos Penetradores de Células/química , Células-Tronco Mesenquimais/metabolismo , Imagem Molecular , Sondas Moleculares/farmacologia , Sequência de Aminoácidos , Animais , Diferenciação Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Peptídeos Penetradores de Células/farmacologia , Modelos Animais de Doenças , Endocitose/efeitos dos fármacos , Feminino , Corantes Fluorescentes/metabolismo , Humanos , Cinética , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Modelos Biológicos , Dados de Sequência Molecular , Osteogênese/efeitos dos fármacos , Ratos Endogâmicos Lew , Febre Reumática/metabolismo , Febre Reumática/patologia , Proteínas Smad/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação/efeitos dos fármacos
10.
Int J Nanomedicine ; 9: 1153-66, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24648725

RESUMO

Protein-transduction technology has been attempted to deliver macromolecular materials, including protein, nucleic acids, and polymeric drugs, for either diagnosis or therapeutic purposes. Herein, fusion protein composed of an arginine-rich cell-penetrating peptide, termed low-molecular-weight protamine (LMWP), and a transcriptional coactivator with a PDZ-binding motif (TAZ) protein was prepared and applied in combination with biomaterials to increase bone-forming capacity. TAZ has been recently identified as a specific osteogenic stimulating transcriptional coactivator in human mesenchymal stem cell (hMSC) differentiation, while simultaneously blocking adipogenic differentiation. However, TAZ by itself cannot penetrate the cells, and thus needs a transfection tool for translocalization. The LMWP-TAZ fusion proteins were efficiently translocalized into the cytosol of hMSCs. The hMSCs treated with cell-penetrating LMWP-TAZ exhibited increased expression of osteoblastic genes and protein, producing significantly higher quantities of mineralized matrix compared to free TAZ. In contrast, adipogenic differentiation of the hMSCs was blocked by treatment of LMWP-TAZ fusion protein, as reflected by reduced marker-protein expression, adipocyte fatty acid-binding protein 2, and peroxisome proliferator-activated receptor-γ messenger ribonucleic acid levels. LMWP-TAZ was applied in alginate gel for the purpose of localization and controlled release. The LMWP-TAZ fusion protein-loaded alginate gel matrix significantly increased bone formation in rabbit calvarial defects compared with alginate gel matrix mixed with free TAZ protein. The protein transduction of TAZ fused with cell-penetrating LMWP peptide was able selectively to stimulate osteogenesis in vitro and in vivo. Taken together, this fusion protein-transduction technology for osteogenic protein can thus be applied in combination with biomaterials for tissue regeneration and controlled release for tissue-engineering purposes.


Assuntos
Peptídeos Penetradores de Células/administração & dosagem , Sistemas de Liberação de Medicamentos , Osteogênese/efeitos dos fármacos , Protaminas/administração & dosagem , Fatores de Transcrição/administração & dosagem , Aciltransferases , Adipogenia/efeitos dos fármacos , Alginatos/administração & dosagem , Animais , Substitutos Ósseos/administração & dosagem , Diferenciação Celular/efeitos dos fármacos , Peptídeos Penetradores de Células/metabolismo , Expressão Gênica , Ácido Glucurônico/administração & dosagem , Ácidos Hexurônicos/administração & dosagem , Humanos , Masculino , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/fisiologia , Nanomedicina , Osteogênese/genética , Osteogênese/fisiologia , Protaminas/metabolismo , Coelhos , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/metabolismo , Engenharia Tecidual/métodos , Fatores de Transcrição/metabolismo
11.
Biomaterials ; 34(17): 4347-59, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23478036

RESUMO

Stem cell differentiation is modulated by several key molecules, including cytokines, hormones, and engineered peptides. Emerging evidence suggests that microRNA has potential applications in stem cell engineering, such as in osteoblastic differentiation. MicroRNAs (miRNAs) bind to the 3'-untranslated region (UTR) sequence of target mRNA, thereby attenuating protein synthesis. Our goal was to evaluate the delivery of miRNA, i.e., miRNA-29b, to stem cells to promote osteoblastic differentiation because this miRNA is known to target anti-osteogenic factors gene expression. Despite the important role of miRNAs, their application has been limited due to poor cell/tissue penetration. The authors attempted to overcome this limitation by using a cell-penetrating peptide (CPP) carrier. Herein, the arginine-rich CPP, called the lowmolecular weight protamine (LMWP), is the sequence from natural protamine. We worked out the difficult problem to transfect into hMSCs by the complex with LMWP, and then we investigated synthetic double-stranded miR-29b could be induced osteoblast differentiation.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Peptídeos Penetradores de Células/farmacologia , Técnicas de Transferência de Genes , Espaço Intracelular/metabolismo , Células-Tronco Mesenquimais/citologia , MicroRNAs/metabolismo , Osteogênese/efeitos dos fármacos , Sequência de Aminoácidos , Fenômenos Biofísicos/efeitos dos fármacos , Diferenciação Celular/genética , Sobrevivência Celular/efeitos dos fármacos , Peptídeos Penetradores de Células/química , Citometria de Fluxo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Espaço Intracelular/efeitos dos fármacos , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Dados de Sequência Molecular , Osteoblastos/citologia , Osteoblastos/metabolismo , Osteogênese/genética , Tamanho da Partícula , Protaminas/química , Eletricidade Estática
12.
J Biomed Mater Res A ; 101(2): 590-8, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22961875

RESUMO

Dentin sialophosphoprotein (DSPP) has been shown to play a primary role in the formation and growth of hydroxyapatite crystals in an extracellular matrix of hard tissue such as bone and teeth. We hypothesized that the mineralization ability of DSPP might depend on a specific domain within it. Three peptides, which have hydroxyapatite (HA) binding affinity, denoted as mineralization inducing peptide (MIP1, MIP2, and MIP3) were identified from DSPP. The both of MIP2 and MIP3 had HA nucleation activity demonstrated by XRD. Among three MIPs, MIP3 significantly supported the human bone marrow stromal cell differentiation into osteoblastic cells. An immunoblot with antibodies specific for the phosphorylated forms of ERK was conducted with cells treated by MIP3. MIP3 transduced intracellular signals via the ERK pathways and was able to induce osteoblastic differentiation, as seen by high expression of ALP, type 1 collagen, OC, OPN, and Runx2 in accordance with applied MIP3 concentration. The Asp, Glu, and Ser residues in MIP3 play important roles for the affinity of calcium in HA bone mineral. Further animal experiment with MIP3 in combination with hydroxyapatite mineral induced marked new bone formation for 4 weeks at rabbit calvarial defect model. The new bone area was much higher in test group, implying that the peptide modified group had excellent biocompatibility when compared with the unmodified group. Taken together, the MIP from DSPP has potential to enhance mineralization followed by to enhance osteoblastic differentiation and bone regeneration.


Assuntos
Regeneração Óssea/efeitos dos fármacos , Calcificação Fisiológica/efeitos dos fármacos , Proteínas da Matriz Extracelular/química , Peptídeos/farmacologia , Fosfoproteínas/química , Sialoglicoproteínas/química , Sequência de Aminoácidos , Animais , Antraquinonas/metabolismo , Regeneração Óssea/genética , Calcificação Fisiológica/genética , Diferenciação Celular/efeitos dos fármacos , Durapatita/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Implantes Experimentais , Masculino , Dados de Sequência Molecular , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Osteogênese/efeitos dos fármacos , Osteogênese/genética , Peptídeos/química , Coelhos , Sefarose , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Crânio/efeitos dos fármacos , Crânio/patologia
13.
Curr Pharm Biotechnol ; 12(8): 1166-82, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21470141

RESUMO

Anticancer drug delivery has been hindered due to cell membrane permeability and the lack of a selective marker for tumor cells. Cell permeability is related to the bioavailability of drugs and has therefore been considered to be an essential step for achieving therapeutic efficacy. While different types of transporters currently exist, cell penetrating peptides (CPPs) have become one of the most popular and effective tools for intracellular drug delivery. Most of the original CPPs are short peptides with basic residues. The mechanism of CPP cell entry remains to be established; however, the CPPs can deliver any type of molecular cargo including solid nanoparticles. Herein, this paper will discuss the classification of CPPs, the mechanism of cell entry, the application of CPPs in tumor therapy, and recent advances in targeted cell penetration that involve CPPs.


Assuntos
Peptídeos Penetradores de Células/administração & dosagem , Sistemas de Liberação de Medicamentos , Neoplasias/tratamento farmacológico , Sequência de Aminoácidos , Animais , Humanos , Dados de Sequência Molecular
14.
Int J Cancer ; 128(10): 2470-80, 2011 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-20669230

RESUMO

In this study, a cell-penetrating peptide, the transactivating transcriptional factor (TAT) domain from HIV, was linked to a chitosan/doxorubicin (chitosan/DOX) conjugate to form a chitosan/DOX/TAT hybrid. The synthesized chitosan/DOX/TAT conjugate showed a different intracellular distribution pattern from a conjugate without TAT. Unlike both free DOX and the conjugate without TAT, the chitosan/DOX/TAT conjugate was capable of efficient cell entry. The chitosan/DOX/TAT conjugate was found to be highly cytotoxic, with an IC(50) value of approximately 480 nM, 2 times less than that of chitosan/DOX (980 nM). The chitosan/DOX/TAT provided decreases in tumor volume of 77.4 and 57.5% compared to free DOX and chitosan/DOX, respectively, in tumor-bearing mice. Therefore, this study suggests that TAT-mediated chitosan/DOX conjugate delivery is effective in slowing tumor growth.


Assuntos
Quitosana/uso terapêutico , Doxorrubicina/uso terapêutico , Neoplasias Experimentais/tratamento farmacológico , Fatores de Transcrição/uso terapêutico , Animais , Quitosana/farmacocinética , Doxorrubicina/farmacocinética , Eletroforese em Gel de Poliacrilamida , Feminino , Citometria de Fluxo , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Confocal
15.
Biomaterials ; 31(28): 7226-38, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20621352

RESUMO

The presence of heparin binding has been become crucial in exerting growth factor related tissue formation. Receptor-mediated osteoblastic differentiation by bone morphogenetic protein (BMP)-4 and supportive function of its heparin binding has been proposed, direct role of the heparin binding site of BMP-4 on osteogenesis has not yet been fully investigated. If the binding site itself plays role on osteogenesis, the site domain can be useful in bone formation in combination with biomaterial. Herein, we synthesized a peptide sequence corresponding to residues 15-24 of BMP-4 (HBD, RKKNPNCRRH), as potential heparin binding sequence. The HBD peptide-induced ostoegenic differentiation by activating extracellular signal-regulated kinase (ERK1/2), one of the key regulators in hMSC. Also, treatment of cultured hMSCs with heparinase blocked both HBD peptide-induced osteogenic differentiation and GAG chain detection while abolishing the increased phospho-ERK level. These results suggest that the identified heparin binding domain peptide (HBD) stimulated osteoblastic differentiation via interaction with heparin and the ERK signaling. In vivo results further demonstrated that HBD, as a form of complex with alginate gel, was able to induce bone formation in the bone defect.


Assuntos
Proteína Morfogenética Óssea 4/metabolismo , Proteína Morfogenética Óssea 4/farmacologia , Heparina/metabolismo , Osteogênese/efeitos dos fármacos , Peptídeos/metabolismo , Peptídeos/farmacologia , Sequência de Aminoácidos , Animais , Biomarcadores/metabolismo , Proteína Morfogenética Óssea 4/genética , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Inibidores Enzimáticos/farmacologia , Heparina/genética , Heparina Liase/metabolismo , Humanos , Masculino , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/fisiologia , Dados de Sequência Molecular , Osteoblastos/citologia , Osteoblastos/fisiologia , Peptídeos/síntese química , Peptídeos/genética , Estrutura Terciária de Proteína , Coelhos , Transdução de Sinais/efeitos dos fármacos
16.
Biomaterials ; 31(6): 1429-43, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19954842

RESUMO

Small interfering RNAs (siRNAs), used for specific down-regulation of targeted genes, have garnered considerable interest as an attractive new class of drugs for broad clinical applications. The polyanionic charges carried by these siRNAs, however, restrain cellular uptake and consequently limit effects on gene regulation. Herein the authors describe a peptide/siRNA complex containing the cell penetrating peptide derived from natural protamine, termed low molecular weight protamine (LMWP), for the treatment of cancer. Fluorescently-tagged siRNAs were localized with the peptide in the cytoplasm shortly after incubation of LMWP/siRNA complex with carcinoma cells. The increased cell uptake of siRNA that was achieved using the LMWP resulted in significant down-regulation of model protein luciferase as well as therapeutic cancer target, vascular endothelial growth factor (VEGF) expression. In vivo studies with tumor-bearing mice further demonstrated that the peptide could carry and localize siRNA inside tumors and inhibit the expression of VEGF through systemic application of the peptide complex, thereby suppressing tumor growth. In addition, no detectable increase in the serum level of inflammatory cytokines including interferon (IFN)-alpha and interleukin (IL)-12 was observed under the LMWP/siRNA complex treatment, indicating systemic delivery of LMWP/siRNA did not exert measurable immunostimulatory effect. The LMWP-based systemic delivery method could be a reliable and safe approach to maximize effectiveness of therapeutic siRNA for treatment of cancer and other diseases.


Assuntos
Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/terapia , Portadores de Fármacos/química , Peptídeos/química , Protaminas/química , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Animais , Inativação Gênica , Terapia Genética/métodos , Masculino , Camundongos , Camundongos Nus , Peso Molecular , Resultado do Tratamento
17.
Biomaterials ; 30(21): 3532-41, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19345406

RESUMO

Self-assembled nanostructures consisting of BMP receptor-binding peptides, termed osteopromotive domains (OPDs), and hydrophobic alkyl chains were fabricated with the aim of developing three-dimensional scaffolding materials for osteoblastic differentiation. OPD peptide was identified from BMP-2 and had an affinity for BMP receptors thereby inducing differentiation of human bone marrow stromal cells into osteoblastic cells. The peptide-hydrophobic alkyl chain amphiphiles were designed to mimic nanofibrous extracellular structures and to add osteogenic ligands to enhance osteoblastic cell function. The OPD peptide-amphiphiles (OPDAs) that end with the alkylation of the N-terminus of the OPD peptide were synthesized by standard solid phase chemistry. The self-assembly was triggered by mixing OPDA solution with calcium ions. Observation using scanning electron microscopy (SEM) revealed the formation of nanofibrous structures with extremely high aspect ratios and high surface areas. The FT-IR and circular dichroism (CD) spectrophotometry demonstrated that self-assembled nanofibers have a beta-sheet structure. The activation of Smad, an osteoblastic differentiation marker, was obtained in the cell culture gel of self-assembled OPDA; therefore, the intracellular signal transduction for osteogenesis was performed like an OPD peptide. Cell survival was supported in the OPDA gel for 10 days, and osteoblastic differentiation of human bone marrow stromal cells (hBMSCs) was evident as demonstrated by calcein staining and ALP activity measurement. These results revealed that self-assembled OPDA maintained osteogenic activity by the surface-exposed OPD peptide. Taken together, the self-assembled OPDA nanofibrous gel can be utilized as a cell culture scaffold in bone regeneration.


Assuntos
Células da Medula Óssea/citologia , Proteína Morfogenética Óssea 2/química , Diferenciação Celular/efeitos dos fármacos , Osteoblastos/citologia , Peptídeos/metabolismo , Peptídeos/farmacologia , Células Estromais/citologia , Materiais Biocompatíveis/síntese química , Materiais Biocompatíveis/química , Materiais Biocompatíveis/metabolismo , Materiais Biocompatíveis/farmacologia , Western Blotting , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/ultraestrutura , Sobrevivência Celular/efeitos dos fármacos , Dicroísmo Circular , Humanos , Microscopia Eletrônica de Varredura , Osteoblastos/ultraestrutura , Peptídeos/síntese química , Peptídeos/química , Espectroscopia de Infravermelho com Transformada de Fourier , Células Estromais/efeitos dos fármacos , Células Estromais/ultraestrutura
18.
Biochem Biophys Res Commun ; 379(3): 669-75, 2009 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-19101509

RESUMO

For the purpose of successfully monitoring labeled cells, optimum labeling efficiency without any side effect is a prerequisite. Magnetic cellular imaging is a new and growing field that allows the visualization of implanted cells in vivo. Herein, superparamagnetic iron oxide (SPIO) nanoparticles were conjugated with a non-toxic protein transduction domain (PTD), identified by the authors and termed low molecular weight protamine (LMWP), to generate efficient and non-toxic cell labeling tools. The cells labeled with LMWP-SPIO presented the highest iron content compared to those labeled with naked SPIO and the complex of SPIO with poly-L-lysine, which is currently used as a transfection agent. In addition to the iron content assay, Prussian staining and confocal observation demonstrated the highest intracellular LMWP-SPIO presence, and the labeling procedure did not alter the cell differentiation capacity of mesenchymal stem cells. Taken together, cell permeable magnetic nanoparticles conjugated with LMWP can be suggested as labeling tools for efficient magnetic imaging of transplanted cells.


Assuntos
Permeabilidade da Membrana Celular , Compostos Férricos/química , Células-Tronco Mesenquimais/metabolismo , Nanopartículas/química , Coloração e Rotulagem/métodos , Linhagem Celular , Coloides/química , Humanos , Magnetismo , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/química , Peptídeos/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA