Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Tissue Eng Regen Med ; 20(6): 965-979, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37589886

RESUMO

BACKGROUND: Inflammatory bowel disease (IBD) is an incurable disease that negatively influences the quality of life of patients. Current and emerging therapies target proinflammatory cytokines and/or receptors to downregulate proinflammatory responses, but insufficient remission requires other therapeutic agents. Herein, we report that the synthetic anti-inflammatory peptide 15 (SAP15) is capable of cell penetration and anti-inflammatory activity in human macrophages. METHODS: SAP15 was labeled with fluorescence and administered to human leukemia monocytic cells (THP-1) cells for cell penetration analysis. Using biolayer interferometry analysis, the binding affinity of SAP15 with histone deacetylase 5 (HDAC5) was measured. SAP15-treated THP-1 cells were analyzed by protein phosphorylation assay, flow cytometry, and enzyme-linked immunosorbent assay (ELISA). In addition, in vivo analysis of the therapeutic effect on IBD was observed in a dextran sulfate sodium (DSS)-induced model. Samples from SAP15-treated mice were analyzed at both the macroscopic and microscopic levels using ELISA, myeloperoxidase (MPO) assays, and histological evaluations. RESULTS: SAP15 was internalized within the cytosol and nucleus of THP-1 cells and bound to the HDAC5 protein. SAP15-treated macrophages were assessed for protein phosphorylation and showed inhibited phosphorylation of HDAC5 and other immune-related proteins, which led to increased M2-like macrophage markers and decreased M1-like macrophage markers and tumor necrosis factor-α and interleukin-6 cytokine levels. The SAP15 treatment on IBD model showed significant recovery of colon length. Further histological analysis of colon demonstrated the therapeutic effect of SAP15 on mucosal layer. Moreover, proinflammatory cytokine levels and MPO activity from the plasma show that SAP15 is effective in reduced proinflammatory responses. CONCLUSION: These findings suggest that SAP15 is a novel peptide with a novel cell-penetrating peptide with anti-inflammatory property that can be used as a therapeutic agent for IBD and other inflammatory diseases.


Assuntos
Peptídeos Penetradores de Células , Doenças Inflamatórias Intestinais , Humanos , Animais , Camundongos , Peptídeos Penetradores de Células/efeitos adversos , Qualidade de Vida , Doenças Inflamatórias Intestinais/tratamento farmacológico , Doenças Inflamatórias Intestinais/induzido quimicamente , Doenças Inflamatórias Intestinais/patologia , Citocinas/metabolismo , Anti-Inflamatórios/farmacologia , Histona Desacetilases/efeitos adversos
2.
Cancers (Basel) ; 15(9)2023 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-37173940

RESUMO

Multiple myeloma (MM) is the second most common hematological malignancy. It is a clonal B-cell disorder characterized by the proliferation of malignant plasma cells in the bone marrow, the presence of monoclonal serum immunoglobulin, and osteolytic lesions. An increasing amount of evidence shows that the interactions of MM cells and the bone microenvironment play a significant role, suggesting that these interactions may be good targets for therapy. The osteopontin-derived collagen-binding motif-bearing peptide NIPEP-OSS stimulates biomineralization and enhances bone remodeling dynamics. Due to its unique targeted osteogenic activity with a broad safety margin, we evaluated the potential of NIPEP-OSS for anti-myeloma activity using MM bone disease (MMBD) animal models. In a 5TGM1-engrafted NSG model, the survival rates of the control and treated groups were significantly different (p = 0.0014), with median survival times of 45 and 57 days, respectively. The bioluminescence analyses showed that myeloma slowly developed in the treated mice compared to the control mice in both models. NIPEP-OSS enhanced bone formation by increasing biomineralization in the bone. We also tested NIPEP-OSS in a well-established 5TGM1-engrafted C57BL/KaLwRij model. Similar to the previous model, the median survival times of the control and treated groups were significantly different (p = 0.0057), with 46 and 63 days, respectively. In comparison with the control, an increase in p1NP was found in the treated mice. We concluded that NIPEP-OSS delays mouse myeloma progression via bone formation in MMBD mouse models.

3.
Biomaterials ; 278: 121156, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34597900

RESUMO

Controlling the senescence of mesenchymal stem cells (MSCs) is essential for improving the efficacy of MSC-based therapies. Here, a model of MSC senescence was established by replicative subculture in tonsil-derived MSCs (TMSCs) using senescence-associated ß-galactosidase, telomere-length related genes, stemness, and mitochondrial metabolism. Using transcriptomic and proteomic analyses, we identified glucose-regulated protein 78 (GRP78) as a unique MSC senescence marker. With increasing cell passage number, GRP78 gradually translocated from the cell surface and cytosol to the (peri)nuclear region of TMSCs. A gelatin-based hydrogel releasing a sustained, low level of reactive oxygen species (ROS-hydrogel) was used to improve TMSC quiescence and self-renewal. TMSCs expressing cell surface-specific GRP78 (csGRP78+), collected by magnetic sorting, showed better stem cell function and higher mitochondrial metabolism than unsorted cells. Implantation of csGRP78+ cells embedded in ROS-hydrogel in rats with calvarial defects resulted in increased bone regeneration. Thus, csGRP78 is a promising biomarker of senescent TMSCs, and the combined use of csGRP78+ cells and ROS-hydrogel improved the regenerative capacity of TMSCs by regulating GRP78 translocation.


Assuntos
Proteínas de Choque Térmico , Células-Tronco Mesenquimais , Espécies Reativas de Oxigênio , Animais , Glucose , Hidrogéis , Proteínas de Membrana , Osteogênese , Tonsila Palatina , Proteômica , Ratos
4.
J Biomed Mater Res A ; 109(10): 2001-2016, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-33818867

RESUMO

Various biomaterials have been used for bone and cartilage regeneration, and inflammation associated with biomaterial implantation is also increased. A 15-mer synthetic anti-inflammatory peptide (SAP15) was designed from human ß-defensin 3 to penetrate cells and induce intracellular downregulation of inflammation. The downregulation of inflammation was achieved by the binding of SAP15 to intracellular histone deacetylase (HDAC5). SAP15-mediated inhibition of inflammation was examined in vitro and in vivo using murine macrophages, human articular chondrocytes, and a collagen-induced arthritis (CIA) rat model. Surface plasmon resonance and immunoprecipitation assays indicated that SAP15 binds to HDAC5. SAP15 inhibited the lipopolysaccharide (LPS)-induced phosphorylation of intracellular HDAC5 and NF-κB p65 in murine macrophages. SAP15 treatment increased aggrecan and type II collagen expression and decreased osteocalcin expression in LPS-induced chondrocytes. Subcutaneous injection of SAP15-loaded sodium hyaluronic acid (HA) solution significantly decreased hind paw swelling, joint inflammation, and serum cytokine levels in CIA rats compared with the effects of sodium HA solution alone. The SAP15-loaded HA group exhibited preservation of cartilage and bone structure in CIA rat joints. Moreover, a more robust anti-inflammatory effect of the SAP15 loaded HA was observed than that of etanercept (an anti-tumor necrosis factor-alpha [TNF-α] antibody)-loaded HA. These findings suggest that SAP15 has an anti-inflammatory effect that is not controlled by sodium HA and is mediated by inhibiting HDAC5, unlike the anti-inflammatory mechanism of etanercept. These results demonstrate that SAP15 is useful as an inflammatory regulator of biomaterials and can be developed as a therapeutic for the treatment of inflammation.


Assuntos
Anti-Inflamatórios/farmacologia , Peptídeos Penetradores de Células/farmacologia , Espaço Intracelular/efeitos dos fármacos , Engenharia de Proteínas , Sequência de Aminoácidos , Animais , Artrite Experimental/sangue , Artrite Experimental/patologia , Peso Corporal/efeitos dos fármacos , Reabsorção Óssea/diagnóstico por imagem , Reabsorção Óssea/patologia , Peptídeos Penetradores de Células/química , Condrócitos/efeitos dos fármacos , Feminino , Histona Desacetilases/metabolismo , Humanos , Inflamação/patologia , Camundongos , Tamanho do Órgão/efeitos dos fármacos , Estrutura Secundária de Proteína , Células RAW 264.7 , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Microtomografia por Raio-X
5.
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
6.
Artigo em Inglês | MEDLINE | ID: mdl-29023613

RESUMO

The purpose of this study was to evaluate the efficacy and safety of equine-derived bone matrix as a carrier for recombinant human platelet-derived growth factor BB (rhPDGF-BB) versus beta-tricalcium phosphate (ß-TCP) for the treatment of intraosseous periodontal defects in adult patients. This study was performed on 32 adults with advanced periodontal disease. Eligible subjects were randomized in 1:1 ratio into a test (rhPDGF-BB-coated equine-derived bone matrix) or control group (rhPDGF-BB-coated ß-TCP). Probing pocket depth (PD), clinical attachment level (CAL), gingival recession (GR), and defect depth on radiographs were measured at 2 weeks before surgery, on the day of surgery (DOS), and 6 months postsurgery (6MPS). The clinical and radiographic data were analyzed over the test period. Statistically significant PD reductions and CAL gain between baseline and 6MPS and between ODS and 6MPS were seen in both groups (P < .01). No statistically significant differences in PD reduction were found between groups. However, the test group showed significant CAL gain between DOS and 6MPS. The radiographic bone level change was statistically significant compared to baseline (P < .01) in both groups. The results suggested that equine-derived bone matrix is a viable, effective, and safe carrier scaffold for rhPDGF in periodontal defects.


Assuntos
Perda do Osso Alveolar/terapia , Materiais Biocompatíveis/administração & dosagem , Matriz Óssea/transplante , Fosfatos de Cálcio/administração & dosagem , Regeneração Tecidual Guiada Periodontal , Perda da Inserção Periodontal/terapia , Proteínas Proto-Oncogênicas c-sis/administração & dosagem , Adulto , Animais , Becaplermina , Terapia Combinada , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas Recombinantes/administração & dosagem , Resultado do Tratamento
7.
J Mol Med (Berl) ; 95(12): 1315-1325, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28889177

RESUMO

Human beta-defensin-3 (HBD3), which is secreted from cells in the skin, salivary gland, and bone marrow, exhibits antimicrobial and immunomodulatory activities. Its C-terminal end contains a 15-amino acid polypeptide (HBD3-C15) that is known to effectively elicit antimicrobial activity. Recently, certain antimicrobial peptides are known to inhibit osteoclast differentiation and, thus, we investigated whether HBD3-C15 hinders osteoclast differentiation and bone destruction to assess its potential use as an anti-bone resorption agent. HBD3-C15 inhibited the receptor activator of nuclear factor κB ligand (RANKL)-induced osteoclast differentiation and formation of resorption pits. In addition, HBD3-C15 disrupted the formation of RANKL-induced podosome belt which is a feature typically found in mature osteoclasts with bone-resorbing capacity. HBD3-C15 downregulated cortactin, cofilin, and vinculin, which are involved in the podosome belt formation. Furthermore, bone loss induced by RANKL was significantly reduced in a mouse calvarial implantation model that was treated with HBD3-C15. Similar inhibitory effects were observed on the osteoclast differentiation and podosome belt formation induced by Aggregatibacter actinomycetemcomitans lipopolysaccharide (AaLPS). Concordantly, HBD3-C15 attenuated the resorption in the calvarial bone of AaLPS-implanted mouse. Collectively, these results suggest that HBD3-C15 has an anti-bone resorption effect in developing osteoclasts and that this occurs via its disruption of podosome belt formation. HBD3-C15 could be a potential therapeutic agent for the inhibition of bone destruction. KEY MESSAGES: HBD3-C15 inhibits osteoclast differentiation and bone resorption capacity. HBD3-C15 disrupts the podosome belt formation in osteoclasts. HBD3-C15 alleviates the bone loss by RANKL or A. actinomycetemcomitans LPS in vivo.


Assuntos
Reabsorção Óssea/tratamento farmacológico , Diferenciação Celular , Osteoclastos/patologia , Peptídeos/uso terapêutico , Podossomos/metabolismo , beta-Defensinas/química , Aggregatibacter/química , Animais , Reabsorção Óssea/patologia , Diferenciação Celular/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Lipopolissacarídeos , Camundongos Endogâmicos C57BL , Fatores de Transcrição NFATC/metabolismo , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Peptídeos/farmacologia , Podossomos/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ligante RANK/farmacologia
8.
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
9.
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
10.
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
11.
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
12.
J Periodontal Implant Sci ; 44(4): 194-200, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25177521

RESUMO

PURPOSE: THE PURPOSE OF THIS STUDY WAS TO ASSESS AND COMPARE THE CLINICAL AND RADIOGRAPHIC OUTCOMES OF GUIDED TISSUE REGENERATION THERAPY FOR HUMAN PERIODONTAL INTRABONY DEFECTS USING TWO DIFFERENT COLLAGEN MEMBRANES: a porous nonchemical cross-linking collagen membrane (NC) and a bilayer collagen membrane (BC). METHODS: Thirty subjects were randomly assigned and divided into the following 3 groups: a test group (NC+BM), in which a NC was used with xenograft bone mineral (BM), a positive control group (BC+BM), in which a BC was used with xenograft BM, and a negative control group (BM), in which only xenograft BM was used. The following clinical measurements were taken at baseline and 3 months after surgery: plaque index, gingival index, probing pocket depth, gingival recession, and clinical attachment level. Radiographic analysis was performed at baseline, 1 week and 3 months after surgery. RESULTS: Membrane exposure was not observed in any cases. Significant probing depth reduction, attachment-level gain and bone fill were observed for both test and control groups compared to baseline at 3 months after surgery (P<0.05). However, there were no statistically significant differences in clinical improvement and radiographic bone fill between treatment protocols (P>0.05). CONCLUSIONS: Within the limitations of this study, the results suggest that both NC and BC were comparable in terms of clinical and radiographic outcomes for the treatment of periodontal intrabony defects in human subjects.

13.
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
14.
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
15.
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
16.
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
17.
J Biomed Mater Res A ; 101(2): 547-54, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22926956

RESUMO

Bone sialoprotein (BSP) is a mineralized, tissue-specific, and noncollagenous protein. The binding of BSP to collagen is thought to be important for the initiation of bone mineralization and formation. In this study, we elucidated the osteogenic efficiency of the collagen-binding (CB) peptide derived from BSP in vitro and in vivo. The CB peptide increased osteoblastic differentiation marker gene and protein expression without affecting cell proliferation. The osteoblastic differentiation by the CB peptide is performed by the activation of extracellular signal-regulated kinase (ERK1/2) and protein kinase B (Akt). Notably, the activation of CB peptide-induced osteogenic differentiation was completely blocked to the basal level by the specific inhibitors for ERK1/2 (U0126) and Akt (LY294002). In vivo results further demonstrated that the CB peptide-coated hydroxyapatite scaffold was able to induce bone formation in the bone defect. Taken together, the CB peptide can be developed as an osteoblastic differentiation agent as well as a fusion biomaterial for bone regeneration therapy.


Assuntos
Colágeno Tipo I/metabolismo , Sialoproteína de Ligação à Integrina/química , Osteogênese/efeitos dos fármacos , Peptídeos/farmacologia , Sequência de Aminoácidos , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Proliferação de Células/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Dados de Sequência Molecular , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Osteogênese/genética , Peptídeos/química , Peptídeos/metabolismo , Ligação Proteica/efeitos dos fármacos , Coelhos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
18.
Int J Nanomedicine ; 7: 5091-106, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23049256

RESUMO

BACKGROUND: Human dental pulp stem cells (DPSCs) have potential applications in tissue regeneration because of their convenient cell harvesting procedures and multipotent capacity. However, the tissue regenerative potential of DPSCs is known to be negatively regulated by aging in long-term culture and under oxidative stress. With an aim of reducing cellular senescence and oxidative stress in DPSCs, an intracellular delivery system for superoxide dismutase 1 (SOD1) was developed. We conjugated SOD1 with a cell-penetrating peptide known as low-molecular weight protamine (LMWP), and investigated the effect of LMWP-SOD1 conjugates on hydrogen peroxide-induced cellular senescence and osteoblastic differentiation. RESULTS: LMWP-SOD1 significantly attenuated enlarged and flattened cell morphology and increased senescence-associated ß-galactosidase activity. Under the same conditions, LMWP-SOD1 abolished activation of the cell cycle regulator proteins, p53 and p21(Cip1), induced by hydrogen peroxide. In addition, LMWP-SOD1 reversed the inhibition of osteoblastic differentiation and downregulation of osteogenic gene markers induced by hydrogen peroxide. However, LMWP-SOD1 could not reverse the decrease in odontogenesis caused by hydrogen peroxide. CONCLUSION: Overall, cell-penetrating LMWP-SOD1 conjugates are effective for attenuation of cellular senescence and reversal of osteoblastic differentiation of DPSCs caused by oxidative stress inhibition. This result suggests potential application in the field of antiaging and tissue engineering to overcome the limitations of senescent stem cells.


Assuntos
Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Polpa Dentária/citologia , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Células-Tronco/citologia , Superóxido Dismutase/administração & dosagem , Proteína Supressora de Tumor p53/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Células Cultivadas , Senescência Celular/efeitos dos fármacos , Senescência Celular/fisiologia , Polpa Dentária/efeitos dos fármacos , Humanos , Osteogênese/efeitos dos fármacos , Osteogênese/fisiologia , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Células-Tronco/efeitos dos fármacos
19.
Biochem Biophys Res Commun ; 419(4): 597-604, 2012 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-22342671

RESUMO

A heparin-binding peptide (HBP) sequence from human heparin-binding epidermal growth factor-like growth factor (HB-EGF) was identified and was shown to exhibit cell penetration activity. This cell penetration induced an anti-inflammatory reaction in lipopolysaccharide (LPS)-treated RAW 264.7 macrophages. HBP penetrated the cell membrane during the 10 min treatment and reduced the LPS-induced production of nitric oxide (NO), inducible nitric oxide synthase (iNOS), and cytokines (TNF-α and IL-6) in a concentration-dependent manner. Additionally, HBP inhibited the LPS-induced upregulation of cytokines, including TNF-α and IL-6, and decreased the interstitial infiltration of polymorphonuclear leukocytes in a lung inflammation model. HBP inhibited NF-κB-dependent inflammatory responses by directly blocking the phosphorylation and degradation of IκBα and by subsequently inhibiting the nuclear translocation of the p65 subunit of NF-κB. Taken together, this novel HBP may be potentially useful candidate for anti-inflammatory treatments and can be combined with other drugs of interest to transport attached molecules into cells.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Peptídeos Penetradores de Células/farmacologia , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Sequência de Aminoácidos , Animais , Anti-Inflamatórios não Esteroides/química , Linhagem Celular , Peptídeos Penetradores de Células/química , Fator de Crescimento Semelhante a EGF de Ligação à Heparina , Humanos , Proteínas I-kappa B/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/química , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Dados de Sequência Molecular , Inibidor de NF-kappaB alfa , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Fosforilação , Conformação Proteica , Estrutura Terciária de Proteína , Proteólise , Ratos , Fator de Transcrição RelA/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
20.
Biochem Biophys Res Commun ; 419(2): 326-32, 2012 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-22342728

RESUMO

Bone sialoprotein (BSP) is a mineralized, tissue-specific, non-collagenous protein that is normally expressed only in mineralized tissues such as bone, dentin, cementum, and calcified cartilage, and at sites of new mineral formation. The binding of BSP to collagen is thought to be important for initiating bone mineralization and bone cell adhesion to the mineralized matrix. Several recent studies have isolated stem cells from muscle tissue, but their functional properties are still unclear. In this study, we examined the effects of a synthetic collagen-binding peptide (CBP) on the differentiation efficiency of muscle-derived stem cells (MDSCs). The CBP sequence (NGVFKYRPRYYLYKHAYFYPHLKRFPVQ) corresponds to residues 35-62 of bone sialoprotein (BSP), which are located within the collagen-binding domain in BSP. Interestingly, this synthetic CBP inhibited adipogenic differentiation but increased osteogenic differentiation in MDSCs. The CBP also induced expression of osteoblastic marker proteins, including alkaline phosphatase (ALP), type I collagen, Runt-related transcription factor 2 (Runx2), and osteocalcin; prevented adipogenic differentiation in MDSCs; and down-regulated adipose-specific mRNAs, such as adipocyte protein 2 (aP2) and peroxisome proliferator-activated receptor γ. The CBP increased Extracellular signal-regulated kinases (ERK) 1/2 protein phosphorylation, which is important in lineage determination. These observations suggest that this CBP determines the osteogenic or adipogenic lineage in MDSCs by activating ERK1/2. Taken together, a novel CBP could be a useful candidate for regenerating bone and treating osteoporosis, which result from an imbalance in osteogenesis and adipogenesis differentiation.


Assuntos
Adipogenia/efeitos dos fármacos , Linhagem da Célula , Separação Celular , Sialoproteína de Ligação à Integrina/farmacologia , Mioblastos Esqueléticos/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Peptídeos/farmacologia , Sialoglicoproteínas/farmacologia , Sequência de Aminoácidos , Colágeno/metabolismo , Meios de Cultura/farmacologia , Humanos , Sialoproteína de Ligação à Integrina/química , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Dados de Sequência Molecular , Mioblastos Esqueléticos/citologia , Mioblastos Esqueléticos/metabolismo , Fragmentos de Peptídeos/química , Peptídeos/química , Conformação Proteica , Sialoglicoproteínas/química , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA