Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
1.
Infect Immun ; 91(11): e0031323, 2023 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-37889003

RESUMO

The widespread prevalence of Helicobacter pylori (H. pylori) infection remains a great challenge to human health. The existing vaccines are not ideal for preventing H. pylori infection; thus, exploring highly effective adjuvants may improve the immunoprotective efficacy of H. pylori vaccines. In a previous study, we found that the outer membrane vesicles (OMVs), a type of nanoscale particle spontaneously produced by Gram-negative bacteria, could act as adjuvants to boost the immune responses to vaccine antigens. In this study, we explored the potential application of OMVs as delivery vectors for adjuvant development. We constructed recombinant OMVs containing eukaryotic expression plasmid of cytokines, including interleukin 17A or interferon-γ, and evaluated their function as adjuvants in combination with inactivated whole-cell vaccine (WCV) or UreB as vaccine antigens. Our results showed that recombinant OMVs as adjuvants could induce stronger humoral and mucosal immune responses in mice than wild-type H. pylori OMVs and the cholera toxin (CT) adjuvant. Additionally, the recombinant OMVs significantly promoted Th1/Th2/Th17-type immune responses. Furthermore, the recombinant OMV adjuvant induced more potent clearance of H. pylori than CT and wild-type OMVs. Our findings suggest that the recombinant OMVs coupled with cytokines may become potent adjuvants for the development of novel and effective vaccines against H. pylori infection.


Assuntos
Infecções por Helicobacter , Helicobacter pylori , Vacinas , Humanos , Animais , Camundongos , Helicobacter pylori/genética , Helicobacter pylori/metabolismo , Citocinas/metabolismo , Infecções por Helicobacter/prevenção & controle , Adjuvantes Imunológicos , Toxina da Cólera/genética , Plasmídeos/genética , Vacinas Bacterianas , Anticorpos Antibacterianos
2.
Appl Microbiol Biotechnol ; 106(22): 7377-7386, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36216901

RESUMO

Ferroptosis is a recently discovered modulated cell death mechanism caused by the accumulation of iron-dependent lipid peroxides to toxic levels and plays an important role in tumor immunology and neurology. Recent studies have shown that ferroptosis may play a crucial role in bacterial infection pathogenesis, which may be useful in anti-infection therapies. However, how bacteria enter cells to induce ferroptosis after invading the host immune system remains largely unknown. In addition, the current studies only focus on the relationship between a single bacterial species or genus and host cell ferroptosis, and there is no systematic summary of its regulatory mechanism. Therefore, our review firstly sums up the role of ferroptosis in bacterial infection and its regulatory mechanism, and innovatively speculates on the function and potential mechanism of extracellular vesicles (EVs) in bacterial-induced ferroptosis, in order to provide possible novel directions and ideas for future anti-infection research. KEY POINTS: • Ferroptosis presents a novel mechanism for bacterial host interaction • EVs provide the potential mechanism for bacterial-induced ferroptosis • The relationship of EVs with ferroptosis provides possible directions for future treatment of bacterial infection.


Assuntos
Vesículas Extracelulares , Ferroptose , Vesículas Extracelulares/metabolismo , Morte Celular , Ferro/metabolismo , Bactérias/metabolismo
3.
FASEB J ; 32(1): 52-62, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28860152

RESUMO

Bone atrophy and its related fragility fractures are frequent, late side effects of radiotherapy in cancer survivors and have a detrimental impact on their quality of life. In another study, we showed that parathyroid hormone 1-34 and anti-sclerostin antibody attenuates radiation-induced bone damage by accelerating DNA repair in osteoblasts. DNA damage responses are partially regulated by the ubiquitin proteasome pathway. In the current study, we examined whether proteasome inhibitors have similar bone-protective effects against radiation damage. MG132 treatment greatly reduced radiation-induced apoptosis in cultured osteoblastic cells. This survival effect was owing to accelerated DNA repair as revealed by γH2AX foci and comet assays and to the up-regulation of Ku70 and DNA-dependent protein kinase, catalytic subunit, essential DNA repair proteins in the nonhomologous end-joining pathway. Administration of bortezomib (Bzb) reversed the loss of trabecular bone structure and strength in mice at 4 wk after focal radiation. Histomorphometry revealed that Bzb significantly increased the number of osteoblasts and activity in the irradiated area and suppressed the number and activity of osteoclasts, regardless of irradiation. Two weeks of Bzb treatment accelerated DNA repair in bone-lining osteoblasts and thus promoted their survival. Meanwhile, it also inhibited bone marrow adiposity. Taken together, we demonstrate a novel role of proteasome inhibitors in treating radiation-induced osteoporosis.-Chandra, A., Wang, L., Young, T., Zhong, L., Tseng, W.-J., Levine, M. A., Cengel, K., Liu, X. S., Zhang, Y., Pignolo, R. J., Qin, L. Proteasome inhibitor bortezomib is a novel therapeutic agent for focal radiation-induced osteoporosis.


Assuntos
Bortezomib/farmacologia , Osteoporose/tratamento farmacológico , Inibidores de Proteassoma/farmacologia , Lesões por Radiação/tratamento farmacológico , Protetores contra Radiação/farmacologia , Células 3T3 , Animais , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Remodelação Óssea/efeitos dos fármacos , Remodelação Óssea/efeitos da radiação , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Osteoblastos/efeitos dos fármacos , Osteoblastos/patologia , Osteoblastos/efeitos da radiação , Osteoporose/metabolismo , Osteoporose/patologia , Lesões por Radiação/metabolismo , Lesões por Radiação/patologia , Microtomografia por Raio-X
4.
Mol Ther ; 26(1): 199-207, 2018 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-28988712

RESUMO

Repair and regeneration of inflammation-induced bone loss remains a clinical challenge. LL37, an antimicrobial peptide, plays critical roles in cell migration, cytokine production, apoptosis, and angiogenesis. Migration of stem cells to the affected site and promotion of vascularization are essential for tissue engineering therapy, including bone regeneration. However, it is largely unknown whether LL37 affects mesenchymal stem cell (MSC) behavior and bone morphogenetic protein 2 (BMP2)-mediated bone repair during the bone pathologic remodeling process. By performing in vitro and in vivo studies with MSCs and a lipopolysaccharide (LPS)-induced mouse calvarial osteolytic bone defect model, we found that LL37 significantly promotes cell differentiation, migration, and proliferation in both unmodified MSCs and BMP2 gene-modified MSCs. Additionally, LL37 inhibited LPS-induced osteoclast formation and bacterial activity in vitro. Furthermore, the combination of LL37 and BMP2 markedly promoted MSC-mediated angiogenesis and bone repair and regeneration in LPS-induced osteolytic defects in mouse calvaria. These findings demonstrate for the first time that LL37 can be a potential candidate drug for promoting osteogenesis and for inhibiting bacterial growth and osteoclastogenesis, and that the combination of BMP2 and LL37 is ideal for MSC-mediated bone regeneration, especially for inflammation-induced bone loss.


Assuntos
Peptídeos Catiônicos Antimicrobianos/farmacologia , Proteína Morfogenética Óssea 2/metabolismo , Regeneração Óssea , Células-Tronco Mesenquimais/metabolismo , Crânio/fisiologia , Animais , Biomarcadores , Proteína Morfogenética Óssea 2/farmacologia , Diferenciação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Lipopolissacarídeos , Camundongos , Osteoclastos/metabolismo , Osteogênese , Osteólise
5.
J Biol Chem ; 290(1): 157-67, 2015 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-25336648

RESUMO

Focal radiotherapy for cancer patients has detrimental effects on bones within the radiation field and the primary clinical signs of bone damage include the loss of functional osteoblasts. We reported previously that daily injection of parathyroid hormone (PTH, 1-34) alleviates radiation-induced osteopenia in a preclinical radiotherapy model by improving osteoblast survival. To elucidate the molecular mechanisms, we irradiated osteoblastic UMR 106-01 cells and calvarial organ culture and demonstrated an anti-apoptosis effect of PTH1-34 on these cultures. Inhibitor assay indicated that PTH exerts its radioprotective action mainly through protein kinase A/ß-catenin pathway. γ-H2AX foci staining and comet assay revealed that PTH efficiently promotes the repair of DNA double strand breaks (DSBs) in irradiated osteoblasts via activating the ß-catenin pathway. Interestingly, Wnt3a alone also blocked cell death and accelerated DNA repair in primary osteoprogenitors, osteoblastic and osteocytic cells after radiation through the canonical signaling. Further investigations revealed that both Wnt3a and PTH increase the amount of Ku70, a core protein for initiating the assembly of DSB repair machinery, in osteoblasts after radiation. Moreover, down-regulation of Ku70 by siRNA abrogated the prosurvival effect of PTH and Wnt3a on irradiated osteoblasts. In summary, our results identify a novel role of PTH and canonical Wnt signaling in regulating DSB repair machinery and apoptosis in osteoblasts and shed light on using PTH1-34 or Wnt agonist as possible therapy for radiation-induced osteoporosis.


Assuntos
Apoptose/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , Osteoblastos/efeitos dos fármacos , Hormônio Paratireóideo/farmacologia , Protetores contra Radiação/farmacologia , Animais , Animais Recém-Nascidos , Antígenos Nucleares/genética , Antígenos Nucleares/metabolismo , Apoptose/efeitos da radiação , Diferenciação Celular , Linhagem Celular Tumoral , Proteínas Quinases Dependentes de AMP Cíclico/genética , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Quebras de DNA de Cadeia Dupla/efeitos da radiação , Reparo do DNA/efeitos da radiação , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica , Autoantígeno Ku , Osteoblastos/citologia , Osteoblastos/efeitos da radiação , Osteócitos/citologia , Osteócitos/efeitos dos fármacos , Osteócitos/efeitos da radiação , Ratos , Proteínas Recombinantes/farmacologia , Transdução de Sinais , Crânio/citologia , Crânio/efeitos dos fármacos , Crânio/efeitos da radiação , Técnicas de Cultura de Tecidos , Proteína Wnt3A/metabolismo , Proteína Wnt3A/farmacologia , Raios X , beta Catenina/genética , beta Catenina/metabolismo
6.
J Biol Chem ; 288(20): 14320-14331, 2013 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-23564457

RESUMO

Collagen VI is a ubiquitously expressed extracellular microfibrillar protein. Its most common molecular form is composed of the α1(VI), α2(VI), and α3(VI) collagen α chains encoded by the COL6A1, COL6A2, and COL6A3 genes, respectively. Mutations in any of the three collagen VI genes cause congenital muscular dystrophy types Bethlem and Ullrich as well as intermediate phenotypes characterized by muscle weakness and connective tissue abnormalities. The α3(VI) collagen α chain has much larger N- and C-globular domains than the other two chains. Its most C-terminal domain can be cleaved off after assembly into microfibrils, and the cleavage product has been implicated in tumor angiogenesis and progression. Here we characterize a Col6a3 mutant mouse that expresses a very low level of a non-functional α3(VI) collagen chain. The mutant mice are deficient in extracellular collagen VI microfibrils and exhibit myopathic features, including decreased muscle mass and contractile force. Ultrastructurally abnormal collagen fibrils were observed in tendon, but not cornea, of the mutant mice, indicating a distinct tissue-specific effect of collagen VI on collagen I fibrillogenesis. Overall, the mice lacking normal α3(VI) collagen chains displayed mild musculoskeletal phenotypes similar to mice deficient in the α1(VI) collagen α chain, suggesting that the cleavage product of the α3(VI) collagen does not elicit essential functions in normal growth and development. The Col6a3 mouse mutant lacking functional α3(VI) collagen chains thus serves as an animal model for COL6A3-related muscular dystrophy.


Assuntos
Colágeno Tipo VI/deficiência , Colágeno Tipo VI/genética , Músculo Esquelético/metabolismo , Tendões/metabolismo , Animais , Colágeno Tipo VI/fisiologia , Modelos Animais de Doenças , Matriz Extracelular/metabolismo , Genótipo , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microfibrilas/metabolismo , Músculo Esquelético/fisiopatologia , Mutação , Fenótipo , Tendões/fisiopatologia
7.
Arthritis Rheum ; 65(12): 3107-19, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23982761

RESUMO

OBJECTIVE: While transforming growth factor ß (TGFß) signaling plays a critical role in chondrocyte metabolism, the TGFß signaling pathways and target genes involved in cartilage homeostasis and the development of osteoarthritis (OA) remain unclear. Using an in vitro cell culture method and an in vivo mouse genetic approach, we undertook this study to investigate TGFß signaling in chondrocytes and to determine whether Mmp13 and Adamts5 are critical downstream target genes of TGFß signaling. METHODS: TGFß receptor type II (TGFßRII)-conditional knockout (KO) (TGFßRII(Col2ER)) mice were generated by breeding TGFßRII(flox/flox) mice with Col2-CreER-transgenic mice. Histologic, histomorphometric, and gene expression analyses were performed. In vitro TGFß signaling studies were performed using chondrogenic rat chondrosarcoma cells. To determine whether Mmp13 and Adamts5 are critical downstream target genes of TGFß signaling, TGFßRII/matrix metalloproteinase 13 (MMP-13)- and TGFßRII/ADAMTS-5-double-KO mice were generated and analyzed. RESULTS: Inhibition of TGFß signaling (deletion of the Tgfbr2 gene in chondrocytes) resulted in up-regulation of Runx2, Mmp13, and Adamts5 expression in articular cartilage tissue and progressive OA development in TGFßRII(Col2ER) mice. Deletion of the Mmp13 or Adamts5 gene significantly ameliorated the OA-like phenotype induced by the loss of TGFß signaling. Treatment of TGFßRII(Col2ER) mice with an MMP-13 inhibitor also slowed OA progression. CONCLUSION: Mmp13 and Adamts5 are critical downstream target genes involved in the TGFß signaling pathway during the development of OA.


Assuntos
Cartilagem Articular/metabolismo , Condrócitos/metabolismo , Osteoartrite/genética , Proteínas Serina-Treonina Quinases/genética , Receptores de Fatores de Crescimento Transformadores beta/genética , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Proteína ADAMTS5 , Animais , Cartilagem Articular/patologia , Condrócitos/patologia , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Progressão da Doença , Metaloproteinase 13 da Matriz/genética , Metaloproteinase 13 da Matriz/metabolismo , Camundongos , Camundongos Knockout , Osteoartrite/metabolismo , Osteoartrite/patologia , Fenótipo , Proteínas Serina-Treonina Quinases/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais , Regulação para Cima
8.
Artigo em Inglês | MEDLINE | ID: mdl-38984547

RESUMO

OBJECTIVES: Back pain subsequent to intervertebral disc (IVD) injury is a common clinical problem. Previous work examining early molecular changes post injury mainly used a candidate marker approach. In this study, gene expression in the injured and intact mouse tail IVDs was determined with a nonbiased whole transcriptome approach. DESIGN: Mouse tail IVD injury was induced by a needle puncture. Whole murine transcriptome was determined by RNASeq. Transcriptomes of injured IVDs were compared with those of intact controls by bioinformatic methods. RESULTS: Among the 18,078 murine genes examined, 592 genes were differentially expressed (P.adj < 0.01). Novel genes upregulated in injured compared with intact IVDs included Chl1, Lum, etc. Ontology study of upregulated genes revealed that leukocyte migration was the most enriched biological process, and network analysis showed that Tnfa had the most protein-protein interactions. Novel downregulated genes in the injured IVDs included 4833412C05Rik, Myoc, etc. The most enriched downregulated pathways were related to cytoskeletal organization. CONCLUSION: Novel genes highly regulated post disc injury were identified with an unbiased approach; they may serve as biomarkers of injury and response to treatments in future experiments. Enriched biological pathways and molecules with high numbers of connections may be targets for treatments post injury.

9.
Am J Phys Med Rehabil ; 103(10): 918-924, 2024 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-38630557

RESUMO

OBJECTIVE: TNFAIP8 and TIPE2 belong to TNFa-induced protein 8 (TNFAIP8/TIPE) family. They control apoptosis and direct leukocyte migration. Nucleus pulposus cell loss is a hallmark of intervertebral disc degeneration in response to injury, and inflammation may cause pain. Here, we examined the effects of TNFAIP8/TIPE2 deficiency on the intervertebral discs in mice with these genes deleted. DESIGN: Tail intervertebral discs in Tnfaip8 or Tipe2 single and double knockout mice ( Tnfaip8 -/- , Tipe2 -/- , and Tnfaip8/Tipe2 dko) , and wild-type controls were injured. The spine motion segments were stained with safranin O to reveal proteoglycans. Macrophages were identified by immunostaining, and selected inflammatory marker and collagen gene expression was examined by Real Time PCR. RESULTS: The injured tail intervertebral discs of Tnfaip -/- , Tipe2 -/- , and Tnfaip8/Tipe2 dko mice all displayed higher levels of proteoglycans than wild-type controls. Fewer macrophages were found in the injured intervertebral discs of Tipe2 -/- and Tnfaip8/Tipe2 dko mice than wild type. Il6 , Adam8 , and Col1 gene expression was downregulated in the injured intervertebral discs of Tnfip8/Tipe2 dko mice. CONCLUSIONS: TNFAIP8 and TIPE2 loss of function ameliorated proteoglycan loss and inflammation in the injured intervertebral discs. They may serve as molecular targets to preserve disc structure and reduce inflammation.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular , Camundongos Knockout , Proteoglicanas , Animais , Camundongos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteoglicanas/metabolismo , Proteoglicanas/genética , Deleção de Genes , Disco Intervertebral/metabolismo , Disco Intervertebral/patologia , Inflamação , Degeneração do Disco Intervertebral/genética , Degeneração do Disco Intervertebral/metabolismo , Degeneração do Disco Intervertebral/patologia , Macrófagos/metabolismo , Modelos Animais de Doenças , Proteínas Reguladoras de Apoptose
10.
J Bone Miner Res ; 39(2): 161-176, 2024 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-38477740

RESUMO

Osteoarthritis (OA) affects multiple tissues in the knee joint, including the synovium and intra-articular adipose tissue (IAAT) that are attached to each other. However, whether these two tissues share the same progenitor cells and hence function as a single unit in joint homeostasis and diseases is largely unknown. Single-cell transcriptomic profiling of synovium and infrapatellar fat pad (IFP), the largest IAAT, from control and OA mice revealed five mesenchymal clusters and predicted mesenchymal progenitor cells (MPCs) as the common progenitors for other cells: synovial lining fibroblasts (SLFs), myofibroblasts (MFs), and preadipocytes 1 and 2. Histologic examination of joints in reporter mice having Dpp4-CreER and Prg4-CreER that label MPCs and SLFs, respectively, demonstrated that Dpp4+ MPCs reside in the synovial sublining layer and give rise to Prg4+ SLFs and Perilipin+ adipocytes during growth and OA progression. After OA injury, both MPCs and SLFs gave rise to MFs, which remained in the thickened synovium at later stages of OA. In culture, Dpp4+ MPCs possessed mesenchymal progenitor properties, such as proliferation and multilineage differentiation. In contrast, Prg4+ SLFs did not contribute to adipocytes in IFP and Prg4+ cells barely grew in vitro. Taken together, we demonstrate that the synovium and joint fat pad are one integrated functional tissue sharing common mesenchymal progenitors and undergoing coordinated changes during OA progression.


Both synovium and intra-articular adipose tissue (IAAT) in knee joint play a critical role in joint health and osteoarthritis (OA) progression. Recent single-cell RNA-sequencing studies have been performed on the mouse and human synovium. However, IAATs residing in close proximity to the synovium have not been studied yet. Our study reveals mesenchymal cell heterogeneity of synovium/infrapatellar fat pad (Syn/IFP) tissue and their OA responses. We identify Dpp4+ multipotent progenitors as a source that give rise to Prg4+ lining layer fibroblasts in the synovium, adipocytes in the IFP, and myofibroblasts in the OA Syn/IFP tissue. Our work demonstrates that Syn/IFP is a functionally connected tissue that shares common mesenchymal progenitors and undergoes coordinated OA changes. This novel insight advances our knowledge of previously understudied joint tissues and provides new directions for drug discovery to treat joint disorders.


Assuntos
Tecido Adiposo , Células-Tronco Mesenquimais , Membrana Sinovial , Animais , Membrana Sinovial/patologia , Membrana Sinovial/metabolismo , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/patologia , Tecido Adiposo/metabolismo , Tecido Adiposo/patologia , Camundongos , Osteoartrite/patologia , Osteoartrite/metabolismo , Patela/patologia , Patela/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA