Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
1.
J Gastroenterol ; 59(8): 744-757, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38727823

RESUMO

BACKGROUND: Chronic pancreatitis (CP) is a progressive disease characterized by pancreatic fibrosis for which effective treatment options are lacking. Mesenchymal stem cells (MSCs) have shown potential for fibrosis treatment but face limitations in clinical application. The high-mobility group box 1 (HMGB1) fragment mobilizes MSCs from bone marrow into the blood and has emerged as a promising therapeutic agent for tissue regeneration in various pathological conditions. The aim of this study was to investigate the potential therapeutic effects of systemic administration of the HMGB1 fragment in a mouse model of CP. METHODS: A caerulein-induced CP mouse model was used, and the HMGB1 fragment was administered by tail vein injection. Parameters such as body weight, pancreatic tissue damage, fibrosis, inflammatory cytokine expression, and collagen-related gene expression were evaluated using various assays, including immunohistochemistry, real-time PCR, serum analysis, and single-cell transcriptome analysis. And the migration of MSCs to the pancreas was evaluated using the parabiosis model. RESULTS: Administration of the HMGB1 fragment was associated with significant improvements in pancreatic tissue damage and fibrosis. It suppressed the expression of inflammatory cytokines and activated platelet-derived growth factor receptor-α+ MSCs, leading to their accumulation in the pancreas. The HMGB1 fragment also shifted gene expression patterns associated with pancreatic fibrosis toward those of the normal pancreas. Systemic administration of the HMGB1 fragment demonstrated therapeutic efficacy in attenuating pancreatic tissue damage and fibrosis in a CP mouse model. CONCLUSION: These findings highlight the potential of the HMGB1 fragment as a therapeutic target for the treatment of CP.


Assuntos
Ceruletídeo , Modelos Animais de Doenças , Fibrose , Proteína HMGB1 , Pâncreas , Pancreatite Crônica , Animais , Pancreatite Crônica/tratamento farmacológico , Proteína HMGB1/metabolismo , Proteína HMGB1/genética , Camundongos , Masculino , Pâncreas/patologia , Camundongos Endogâmicos C57BL , Citocinas/metabolismo , Células-Tronco Mesenquimais/metabolismo , Transplante de Células-Tronco Mesenquimais/métodos
2.
Biochem Biophys Res Commun ; 671: 357-365, 2023 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-37329659

RESUMO

BACKGROUND: This study aimed to examine the effect of the HMGB1 peptide on Bronchopulmonary dysplasia (BPD)-related lung injury in a mouse model. RESULTS: HMGB1 peptide ameliorates lung injury by suppressing the release of inflammatory cytokines and decreasing soluble collagen levels in the lungs. Single-cell RNA sequencing showed that the peptide suppressed the hyperoxia-induced inflammatory signature in macrophages and the fibrotic signature in fibroblasts. These changes in the transcriptome were confirmed using protein assays. CONCLUSION: Systemic administration of HMGB1 peptide exerts anti-inflammatory and anti-fibrotic effects in a mouse model of BPD. This study provides a foundation for the development of new and effective therapies for BPD.


Assuntos
Displasia Broncopulmonar , Proteína HMGB1 , Hiperóxia , Lesão Pulmonar , Animais , Humanos , Camundongos , Recém-Nascido , Displasia Broncopulmonar/tratamento farmacológico , Displasia Broncopulmonar/genética , Lesão Pulmonar/patologia , Proteína HMGB1/metabolismo , Animais Recém-Nascidos , Pulmão/patologia , Hiperóxia/patologia , Citocinas/efeitos adversos , Inflamação/tratamento farmacológico , Inflamação/patologia , Modelos Animais de Doenças , Fibrose
3.
J Invest Dermatol ; 143(12): 2447-2455.e8, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37302620

RESUMO

Recessive dystrophic epidermolysis bullosa (RDEB) is a genodermatosis caused by variants in COL7A1-encoded type VII collagen, a major component of anchoring fibrils. In this study, we developed an ex vivo gene therapy for RDEB using autologous mesenchymal stromal cells (MSCs). On the basis of our previous studies, we first attempted to isolate MSCs from the blister fluid of patients with RDEB and succeeded in obtaining cells with a set of MSC characteristics from all 10 patients. We termed these cells blister fluid-derived MSCs. Blister fluid-derived MSCs were genetically modified and injected into skins of type VII collagen-deficient neonatal mice transplanted onto immunodeficient mice, resulting in continuous and widespread expression of type VII collagen at the dermal-epidermal junction, particularly when administered into blisters. When injected intradermally, the efforts were not successful. The gene-modified blister fluid-derived MSCs could be cultured as cell sheets and applied to the dermis with an efficacy equivalent to that of intrablister administration. In conclusion, we successfully developed a minimally invasive and highly efficient ex vivo gene therapy for RDEB. This study shows the successful application of gene therapy in the RDEB mouse model for both early blistering skin and advanced ulcerative lesions.


Assuntos
Epidermólise Bolhosa Distrófica , Células-Tronco Mesenquimais , Humanos , Camundongos , Animais , Epidermólise Bolhosa Distrófica/genética , Epidermólise Bolhosa Distrófica/terapia , Epidermólise Bolhosa Distrófica/patologia , Vesícula/genética , Vesícula/terapia , Colágeno Tipo VII/genética , Colágeno Tipo VII/metabolismo , Pele/patologia , Genes Recessivos , Células-Tronco Mesenquimais/metabolismo
4.
Hepatol Res ; 52(12): 985-997, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35932481

RESUMO

AIM: Non-alcoholic steatohepatitis (NASH) with fibrosis eventually leads to cirrhosis and hepatocellular carcinoma. Thus, the development of therapies other than dietary restriction and exercise, particularly those that suppress steatosis and fibrosis of the liver and have a long-term beneficial effect, is necessary. We aimed to evaluate the therapeutic effects of the HMGB1 peptide synthesized from box A using the melanocortin-4 receptor-deficient (Mc4r-KO) NASH model mouse. METHODS: We performed short- and long-term administration of this peptide and evaluated the effects on steatosis, fibrosis, and carcinogenesis using Mc4r-KO mice. We also analyzed the direct effect of this peptide on macrophages and hepatic stellate cells in vitro and performed lipidomics and metabolomics techniques to evaluate the effect. RESULTS: Although this peptide did not show direct effects on macrophages and hepatic stellate cells in vitro, in the short-term administration model, we could confirm the reduction of liver damage, steatosis, and fibrosis progression. The results of lipidomics and metabolomics suggested that the peptide might ameliorate NASH by promoting lipolysis via the activation of fatty acid ß-oxidation and improving insulin resistance. In the long-term administration model, this peptide prevented progression to cirrhosis but retained the steatosis state, that is, the peptide prevents the progression to "burnt-out NASH." This peptide inhibited carcinogenesis by about one-third. CONCLUSION: This HMGB1 peptide can reduce liver damage, improve fibrosis and steatosis, and inhibit carcinogenesis, suggesting that the peptide would be a new treatment candidate for NASH and can contribute to the long-term prognosis for patients with NASH.

5.
Lasers Surg Med ; 54(8): 1167-1176, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35916125

RESUMO

OBJECTIVES: Hair loss, including alopecia, is a common dermatological issue worldwide. At present, the application of fractional carbon dioxide (CO2 ) laser in the treatment of alopecia has been documented; however, the results vary between reports. These varying results may be due to the limited knowledge of cellular action in laser-irradiated skin. The objective of this study was to investigate the molecular and cellular mechanisms of laser treatment under effective conditions for hair cycle initiation. METHODS: A fractional CO2 laser was applied and optimized to initiate the hair cycle in a mouse model of alopecia. Several cellular markers were analyzed in the irradiated skin using immunofluorescence staining. Cellular populations and their comprehensive gene expression were analyzed using single-cell RNA sequencing and bioinformatics. RESULTS: The effective irradiation condition for initiating the hair cycle was found to be 15 mJ energy/spot, which generates approximately 500 µm depth columns, but does not penetrate the dermis, only reaching approximately 1 spot/mm2 . The proportion of macrophage clusters significantly increased upon irradiation, whereas the proportion of fibroblast clusters decreased. The macrophages strongly expressed C-C chemokine receptor type 2 (Ccr2), which is known to be a key signal for injury-induced hair growth. CONCLUSIONS: We found that fractional CO2 laser irradiation recruited Ccr2 positive macrophages, and induced hair regrowth in a mouse alopecia model. These findings may contribute to the development of stable and effective fractional laser irradiation conditions for human alopecia treatment.


Assuntos
Dióxido de Carbono , Lasers de Gás , Alopecia/genética , Alopecia/radioterapia , Animais , Dióxido de Carbono/farmacologia , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Cabelo , Humanos , Lasers de Gás/uso terapêutico , Camundongos
6.
Eur J Immunol ; 52(2): 204-221, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34708880

RESUMO

Multiple embryonic precursors give rise to leukocytes in adults while the lineage-based functional impacts are underappreciated. Mesodermal precursors expressing PDGFRα appear transiently during E7.5-8.5 descend to a subset of Lin- Sca1+ Kit+ hematopoietic progenitors found in adult BM. By analyzing a PDGFRα-lineage tracing mouse line, we here report that PDGFRα-lineage BM F4/80+ SSClo monocytes/macrophages are solely Ly6C+ LFA-1hi Mac-1hi monocytes enriched on the abluminal sinusoidal endothelium while Ly6C- LFA-1lo Mac-1lo macrophages are mostly from non-PDGFRα-lineage in vivo. Monocytes with stronger integrin profiles outcompete macrophages for adhesion on an endothelial monolayer or surfaces coated with ICAM-1-Fc or VCAM-1-Fc. Egress of PDGFRα-lineage-rich monocytes and subsequent differentiation to peripheral macrophages spatially segregates them from non-PDGFRα-lineage BM-resident macrophages and allows functional specialization since macrophages derived from these egressing monocytes differ in morphology, phenotype, and functionality from BM-resident macrophages in culture. Extravasation preference for blood PDGFRα-lineage monocytes varies by tissues and governs the local lineage composition of macrophages. More PDGFRα-lineage classical monocytes infiltrated into skin and colon but not into peritoneum. Accordingly, transcriptomic analytics indicated augmented inflammatory cascades in dermatitis skin of BM-chimeric mice harbouring only PDGFRα-lineage leukocytes. Thus, the PDGFRα-lineage origin biasedly generates monocytes predestined for BM exit to support peripheral immunity following extravasation and macrophage differentiation.


Assuntos
Linhagem da Célula/imunologia , Movimento Celular/imunologia , Endotélio Vascular/imunologia , Macrófagos/imunologia , Monócitos/imunologia , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/imunologia , Animais , Linhagem da Célula/genética , Movimento Celular/genética , Camundongos , Camundongos Transgênicos , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética
8.
Cell Mol Gastroenterol Hepatol ; 12(2): 547-566, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33862275

RESUMO

BACKGROUND & AIMS: Proper resolution of inflammation is essential to maintaining homeostasis, which is important as a dysregulated inflammatory response has adverse consequences, even being regarded as a hallmark of cancer. However, our picture of dynamic changes during inflammation remains far from comprehensive. METHODS: Here we used single-cell transcriptomics to elucidate changes in distinct cell types and their interactions in a mouse model of chemically induced colitis. RESULTS: Our analysis highlights the stromal cell population of the colon functions as a hub with dynamically changing roles over time. Importantly, we found that Serpina3n, a serine protease inhibitor, is specifically expressed in stromal cell clusters as inflammation resolves, interacting with a potential target, elastase. Indeed, genetic ablation of the Serpina3n gene delays resolution of induced inflammation. Furthermore, systemic Serpina3n administration promoted the resolution of inflammation, ameliorating colitis symptoms. CONCLUSIONS: This study provides a comprehensive, single-cell understanding of cell-cell interactions during colorectal inflammation and reveals a potential therapeutic target that leverages inflammation resolution.


Assuntos
Proteínas de Fase Aguda/metabolismo , Colite/genética , Colite/patologia , Inflamação/genética , Inflamação/patologia , Serpinas/metabolismo , Análise de Célula Única , Transcriptoma/genética , Animais , Comunicação Celular , Colo/patologia , Sulfato de Dextrana , Modelos Animais de Doenças , Predisposição Genética para Doença , Doenças Inflamatórias Intestinais/genética , Doenças Inflamatórias Intestinais/patologia , Camundongos Endogâmicos C57BL , Fenótipo , RNA-Seq , Fatores de Risco , Células Estromais/metabolismo
9.
Biochem Biophys Res Commun ; 539: 83-88, 2021 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-33461067

RESUMO

Feto-maternal immune tolerance is established during pregnancy; however, its mechanism and maintenance remain underexplored. Here, we investigated whether mesenchymal stem/stromal cells (MSCs) as non-inherited maternal antigens (NIMAs) transferred by maternal microchimerism could induce immune tolerance. We showed that MSCs had a potential equivalent to hematopoietic stem and progenitor cells (HSPCs) to induce immune tolerance and that MSCs were essential to induce tolerance to MSC-specific antigens. Furthermore, we demonstrated that MSCs as NIMAs transferred by maternal microchimerism could induce robust immune tolerance that can be further enhanced using a drug. Our data shed light on induction of immune tolerance and serve as a foundation to develop new therapies using maternally derived cells for autoimmune or genetic diseases.


Assuntos
Quimera/imunologia , Células-Tronco Hematopoéticas/imunologia , Troca Materno-Fetal/imunologia , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/imunologia , Animais , Feminino , Células-Tronco Hematopoéticas/citologia , Tolerância Imunológica , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Gravidez
10.
Biochem Biophys Res Commun ; 534: 186-192, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33309273

RESUMO

Platelet-derived growth factor receptor alpha (PDGFRα) is a dominant marker of mesodermal mesenchymal cells in mice. Previous studies demonstrated that PDGFRα-positive (PDGFRα+) mesodermal cells develop not only into mesenchymal cells but also into a subset of total hematopoietic cells (HCs) in the limited period during mouse embryogenesis. However, the precise characteristics of the PDGFRα lineage positive (PDGFRα Lin+) HCs in adult mouse hematopoiesis are largely unknown. In this study, we systematically evaluated the characteristics of PDGFRα Lin+ HCs in the bone marrow and peripheral blood using PDGFRα-CRE; ROSAtdTomato mice. Flow cytometry analysis revealed that PDGFRα Lin+ HCs accounted for approximately 20% of total HCs in both the bone marrow and peripheral blood in adult mice. Compositions of myeloid and lymphoid subpopulations among CD45+ mononuclear cells were almost identical in both PDGFRα Lin+ and PDGFRα Lin- cells. Single-cell RNA-sequencing analysis also demonstrated that the transcriptomic signatures of the PDGFRα Lin+ HCs in the peripheral blood largely overlapped with those of the PDGFRα Lin- HCs, suggesting equivalent functions of the PDGFRα Lin+ and PDGFRα Lin- HCs. Although pathophysiological activities of the PDGFRα Lin + HCs were not evaluated, our data clearly demonstrate a significant role of the PDGFRα Lin + HCs in physiological hematopoiesis in adult mice.


Assuntos
Hematopoese , Células-Tronco Hematopoéticas/fisiologia , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Animais , Linhagem da Célula , Feminino , Hematopoese/genética , Células-Tronco Hematopoéticas/metabolismo , Masculino , Mesoderma/citologia , Camundongos , RNA-Seq , Análise de Célula Única
11.
PLoS One ; 15(4): e0230392, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32275672

RESUMO

OBJECTIVES: High-mobility group box 1 protein (HMGB1) fragment enhances bone marrow-derived mesenchymal stem cell (BM-MSC) recruitment to damaged tissue to promote tissue regeneration. This study aimed to evaluate whether systemic injection of HMGB1 fragment could promote tissue repair in a rat model of myocardial infarction (MI). METHODS: HMGB1 (n = 14) or phosphate buffered saline (n = 12, control) was administered to MI rats for 4 days. Cardiac performance and left ventricular remodeling were evaluated using ultrasonography and immunostaining. BM-MSC recruitment to damaged tissue in green fluorescent protein-bone marrow transplantation (GFP-BMT) models was evaluated using immunostaining. RESULTS: At four weeks post-treatment, the left ventricular ejection fraction was significantly improved in the HMGB1 group compared to that in the control. Interstitial fibrosis and cardiomyocyte hypertrophy were also significantly attenuated in the HMGB1 group compared to the control. In the peri-infarction area, VEGF-A mRNA expression was significantly higher and TGFß expression was significantly attenuated in the HMGB1 group than in the control. In GFP-BMT rats, GFP+/PDGFRα+ cells were significantly mobilized to the peri-infarction area in the HMGB1 group compared to that in the control, leading to the formation of new vasculature. In addition, intravital imaging revealed that more GFP+/PDGFRα+ cells were recruited to the peri-infarction area in the HMGB1 group than in the control 12 h after treatment. CONCLUSIONS: Systemic administration of HMGB1 induced angiogenesis and reduced fibrosis by recruiting PDGFRα+ mesenchymal cells from the bone marrow, suggesting that HMGB1 administration might be a new therapeutic approach for heart failure after MI.


Assuntos
Proteína HMGB1/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Infarto do Miocárdio/tratamento farmacológico , Indutores da Angiogênese/farmacologia , Animais , Modelos Animais de Doenças , Fibrose/tratamento farmacológico , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Coração/fisiopatologia , Insuficiência Cardíaca/tratamento farmacológico , Masculino , Células-Tronco Mesenquimais/metabolismo , Infarto do Miocárdio/fisiopatologia , Ratos , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Regeneração/efeitos dos fármacos
12.
Case Rep Dermatol ; 11(3): 256-259, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31616278

RESUMO

Dermatofibroma (DF) is a benign skin tumor that is well-known among dermatologists. We herein present a rare case of atrophic dermatofibroma presenting linear skin dimpling. The patient was a 25-year-old woman with a history of wild-type recessive dystrophic epidermolysis bullosa who had noticed linear concavity on her right lateral back 1 year before her initial presentation. Anetoderma, atrophic scar, localized morphea, or lupus erythematosus profundus were clinically suspected; however, a biopsy specimen from the dimpling lesion showed the fibrous and histiocytic tumor in the deep dermis. The spindle-to-rhomboid-shaped tumor cells were arranged with irregularly storiform pattern, and immunohistochemistry showed that the tumor cells were positive for factor XIIIa, and negative for CD34 and CD68. Elastica van Gieson staining showed an almost complete loss of elastic fibers, especially at the center of the lesion. The reduction of elastic fibers might have influenced the skin depression in this case. This rare case suggests the need to consider a subtype of DF in the differential diagnosis of dimpling skin lesions.

13.
Biochem Biophys Res Commun ; 512(2): 326-330, 2019 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-30890337

RESUMO

Umbilical cord blood contains mesenchymal stem/stromal cells (MSCs) in addition to hematopoietic stem cells, serving as an attractive tool for regenerative medicine. As umbilical cord blood originates from fetus, abundant MSCs are expected to circulate in fetus. However, the properties of circulating MSCs in fetus have not been fully examined. In the present study, we aimed to analyze circulating MSCs, marked by the expression of platelet-derived growth factor receptor α (PDGFRα), during fetal development. Using PDGFRα GFP knock-in mice, we quantified the number of circulating PDGFRα positive MSCs during development. We further performed whole transcriptome analysis of circulating MSCs at single cell levels. We found that abundant PDGFRα positive cells circulate in embryo and diminish immediately after birth. In addition, single cell RNA-sequencing revealed transcriptional heterogeneity of MSCs in fetal circulation. These data lay a foundation to analyze the function of circulating MSCs during development.


Assuntos
Sangue Fetal/citologia , Sangue Fetal/metabolismo , Feto/citologia , Feto/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Animais , Contagem de Células , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Gravidez , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Medicina Regenerativa , Análise de Célula Única , Transcrição Gênica
14.
Sci Rep ; 8(1): 17765, 2018 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-30531792

RESUMO

Mesenchymal stem cells (MSCs), which can differentiate into tri-lineage (osteoblast, adipocyte, and chondrocyte) and suppress inflammation, are promising tools for regenerative medicine. MSCs are phenotypically diverse based on their tissue origins. However, the mechanisms underlying cell-type-specific gene expression patterns are not fully understood due to the lack of suitable strategy to identify the diversity. In this study, we investigated gene expression programs and chromatin accessibilities of MSCs by whole-transcriptome RNA-seq analysis and an assay for transposase-accessible chromatin using sequencing (ATAC-seq). We isolated MSCs from four tissues (femoral and vertebral bone marrow, adipose tissue, and lung) and analysed their molecular signatures. RNA-seq identified the expression of MSC markers and both RNA-seq and ATAC-seq successfully clustered the MSCs based on their tissue origins. Interestingly, clustering based on tissue origin was more accurate with chromatin accessibility signatures than with transcriptome profiles. Furthermore, we identified transcription factors potentially involved in establishing cell-type specific chromatin structures. Thus, epigenome analysis is useful to analyse MSC identity and can be utilized to characterize these cells for clinical use.


Assuntos
Cromatina/genética , Cromatina/metabolismo , Células-Tronco Mesenquimais/metabolismo , Adipócitos/metabolismo , Adipócitos/fisiologia , Tecido Adiposo/metabolismo , Tecido Adiposo/fisiologia , Animais , Biomarcadores/metabolismo , Células da Medula Óssea/metabolismo , Células da Medula Óssea/fisiologia , Análise por Conglomerados , Fêmur/metabolismo , Fêmur/fisiologia , Expressão Gênica/genética , Expressão Gênica/fisiologia , Perfilação da Expressão Gênica/métodos , Pulmão/metabolismo , Pulmão/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcriptoma/genética , Transcriptoma/fisiologia
15.
PLoS One ; 13(12): e0202838, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30517097

RESUMO

OBJECTIVES: We hypothesized that systemic administration of high-mobility group box 1 fragment attenuates the progression of myocardial fibrosis and cardiac dysfunction in a hamster model of dilated cardiomyopathy by recruiting bone marrow mesenchymal stem cells thus causing enhancement of a self-regeneration system. METHODS: Twenty-week-old J2N-k hamsters, which are δ-sarcoglycan-deficient, were treated with systemic injection of high-mobility group box 1 fragment (HMGB1, n = 15) or phosphate buffered saline (control, n = 11). Echocardiography for left ventricular function, cardiac histology, and molecular biology were analyzed. The life-prolonging effect was assessed separately using the HMGB1 and control groups, in addition to a monthly HMGB1 group which received monthly systemic injections of high-mobility group box 1 fragment, 3 times (HMGB1, n = 11, control, n = 9, monthly HMGB1, n = 9). RESULTS: The HMGB1 group showed improved left ventricular ejection fraction, reduced myocardial fibrosis, and increased capillary density. The number of platelet-derived growth factor receptor-alpha and CD106 positive mesenchymal stem cells detected in the myocardium was significantly increased, and intra-myocardial expression of tumor necrosis factor α stimulating gene 6, hepatic growth factor, and vascular endothelial growth factor were significantly upregulated after high-mobility group box 1 fragment administration. Improved survival was observed in the monthly HMGB1 group compared with the control group. CONCLUSIONS: Systemic high-mobility group box 1 fragment administration attenuates the progression of left ventricular remodeling in a hamster model of dilated cardiomyopathy by enhanced homing of bone marrow mesenchymal stem cells into damaged myocardium, suggesting that high-mobility group box 1 fragment could be a new treatment for dilated cardiomyopathy.


Assuntos
Células da Medula Óssea/metabolismo , Cardiomiopatia Dilatada , Autorrenovação Celular , Proteína HMGB1/farmacologia , Células-Tronco Mesenquimais/metabolismo , Sarcoglicanas/deficiência , Função Ventricular Esquerda , Animais , Animais Geneticamente Modificados , Células da Medula Óssea/patologia , Cardiomiopatia Dilatada/diagnóstico por imagem , Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/metabolismo , Cardiomiopatia Dilatada/fisiopatologia , Autorrenovação Celular/efeitos dos fármacos , Autorrenovação Celular/genética , Cricetinae , Modelos Animais de Doenças , Ecocardiografia , Fibrose , Proteína HMGB1/genética , Células-Tronco Mesenquimais/patologia , Função Ventricular Esquerda/efeitos dos fármacos , Função Ventricular Esquerda/genética
16.
Geriatr Gerontol Int ; 17(11): 2150-2156, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28488306

RESUMO

AIM: In patients with diabetes or ischemia, angiogenesis and infection control are required for chronic leg ulcers, which substantially impair patients' quality of life. We developed a novel functional peptide, named AG30/5C, with angiogenic and anti-microbial properties. Treatment with AG30/5C significantly accelerated the wound healing of full-thickness defects in mice. To evaluate the safety of AG30/5C in the treatment of leg ulcers, a physician-initiated clinical study was carried out. METHODS: The first-in-human trial was designed as an open-label treatment with AG30/5C (0.1 mg/mL) given twice per day for 11 days, and with a follow-up period of 17 days. The inclusion criteria for severe skin ulcers were: (i) diabetes or critical limb ischemia; (ii) resistance to standard therapy for 1 month; and (iii) detection of methicillin-resistant Staphylococcus aureus in the skin ulcer. RESULTS: Four patients were enrolled in this study, and two patients met these criteria. For the evaluation of safety, three adverse effects were reported as possibly related to AG30/5C treatment; however, these adverse effects were not severe and resolved during or after treatment. Thus, there were no safety concerns. In both patients, the size of the ulcer decreased after treatment (44.62% and 10.23% decrease), and further decreased after the follow-up period (73.85% and 10.23% decrease). The former patient was diagnosed as Werner syndrome and the skin ulcer was resistant to standard therapy; however, it was sensitive to AG30/5C treatment. CONCLUSIONS: Topical treatment with AG30/5C for severe leg ulcers was safe, well tolerated and effective. Geriatr Gerontol Int 2017; 17: 2150-2156.


Assuntos
Proteínas Angiogênicas/uso terapêutico , Úlcera da Perna/terapia , Peptídeos/uso terapêutico , Proteínas Angiogênicas/efeitos adversos , Humanos , Peptídeos/efeitos adversos , Médicos , Índice de Gravidade de Doença , Resultado do Tratamento
17.
J Invest Dermatol ; 136(11): 2119-2121, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27772543

RESUMO

Epidermolysis bullosa is a group of heritable skin fragility disorders with considerable morbidity and mortality. It is known to be caused by mutations in as many as 18 distinct genes, but there is no specific or effective treatment. Preclinical developments of gene correction, protein replacement, and cell-based approaches for treatment have suggested new therapeutic avenues, and some of them, including bone marrow transplantation and mesenchymal stem cell therapy, have entered into early clinical trials. Hammersen et al. report on two patients with severe generalized junctional epidermolysis bullosa treated with allogeneic stem cell therapy, but with little success. Careful examination of the existing literature suggests that current approaches of cell-based therapies may be helpful in ameliorating some of the clinical features and symptoms in these patients, but advanced strategies, with improved safety profiles, are required for development of durable therapy for these currently intractable disorders.


Assuntos
Epidermólise Bolhosa/terapia , Terapia Genética/métodos , Transplante de Células-Tronco/métodos , Epidermólise Bolhosa/genética , Humanos
18.
Stem Cells Dev ; 25(22): 1721-1732, 2016 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-27539289

RESUMO

Bone marrow mesenchymal stromal cells (BM-MSCs) have been demonstrated to contribute to tissue regeneration. However, chronic pathological conditions, such as diabetes and aging, can result in a decreased number and/or quality of BM-MSCs. We therefore investigated the maintenance mechanism of BM-MSCs by studying signaling through the receptor for advanced glycation end products (RAGE), which is thought to be activated under various pathological conditions. The abundance of endogenous BM-MSCs decreased in a type 2 diabetes mellitus (DM2) model, as determined by performing colony-forming unit (CFU) assays. Flow cytometric analysis revealed that the prevalence of the Lin-/ckit-/CD106+/CD44- BM population, which was previously identified as a slow-cycling BM-MSC population, also decreased. Furthermore, in a streptozotocin-induced type 1 DM model (DM1), the CFUs of fibroblasts and the prevalence of the Lin-/ckit-/CD106+/CD44- BM population also significantly decreased. BM-MSCs in RAGE knockout (KO) mice were resistant to such reduction induced by streptozotocin treatment, suggesting that chronic RAGE signaling worsened the maintenance mechanism of BM-MSCs. Using an in vitro culture condition, BM-MSCs from RAGE-KO mice showed less proliferation and expressed significantly more Nanog and Oct-4, which are key factors in multipotency, than did wild-type BM-MSCs. Furthermore, RAGE-KO BM-MSCs showed a greater capacity for differentiation into mesenchymal lineages, such as adipocytes and osteocytes. These data suggested that RAGE signaling inhibition is useful for maintaining BM-MSCs in vitro. Together, our findings indicated that perturbation of BM-MSCs in DM could be partially explained by chronic RAGE signaling and that targeting the RAGE signaling pathway is a viable approach for maintaining BM-MSCs under chronic pathological conditions.


Assuntos
Células da Medula Óssea/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Células-Tronco Mesenquimais/metabolismo , Receptor para Produtos Finais de Glicação Avançada/metabolismo , Transdução de Sinais , Animais , Contagem de Células , Diferenciação Celular , Proliferação de Células , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patologia , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL , Camundongos Knockout
19.
PLoS One ; 10(12): e0144166, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26657737

RESUMO

Substances that enhance the migration of mesenchymal stem cells to damaged sites have the potential to improve the effectiveness of tissue repair. We previously found that ethanol extracts of Mallotus philippinensis bark promoted migration of mesenchymal stem cells and improved wound healing in a mouse model. We also demonstrated that bark extracts contain cinnamtannin B-1, a flavonoid with in vitro migratory activity against mesenchymal stem cells. However, the in vivo effects of cinnamtannin B-1 on the migration of mesenchymal stem cells and underlying mechanism of this action remain unknown. Therefore, we examined the effects of cinnamtannin B-1 on in vivo migration of mesenchymal stem cells and wound healing in mice. In addition, we characterized cinnamtannin B-1-induced migration of mesenchymal stem cells pharmacologically and structurally. The mobilization of endogenous mesenchymal stem cells into the blood circulation was enhanced in cinnamtannin B-1-treated mice as shown by flow cytometric analysis of peripheral blood cells. Whole animal imaging analysis using luciferase-expressing mesenchymal stem cells as a tracer revealed that cinnamtannin B-1 increased the homing of mesenchymal stem cells to wounds and accelerated healing in a diabetic mouse model. Additionally, the cinnamtannin B-1-induced migration of mesenchymal stem cells was pharmacologically susceptible to inhibitors of phosphatidylinositol 3-kinase, phospholipase C, lipoxygenase, and purines. Furthermore, biflavonoids with similar structural features to cinnamtannin B-1 also augmented the migration of mesenchymal stem cells by similar pharmacological mechanisms. These results demonstrate that cinnamtannin B-1 promoted mesenchymal stem cell migration in vivo and improved wound healing in mice. Furthermore, the results reveal that cinnamtannin B-1-induced migration of mesenchymal stem cells may be mediated by specific signaling pathways, and the flavonoid skeleton may be relevant to its effects on mesenchymal stem cell migration.


Assuntos
Movimento Celular/efeitos dos fármacos , Células-Tronco Mesenquimais/citologia , Proantocianidinas/farmacologia , Cicatrização/efeitos dos fármacos , Animais , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Diabetes Mellitus Experimental/patologia , Modelos Animais de Doenças , Feminino , Flavonoides/farmacologia , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Camundongos , Polifenóis/farmacologia
20.
Sci Rep ; 5: 11008, 2015 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-26046579

RESUMO

High-mobility group box 1 (HMGB1) mobilizes platelet-derived growth factor receptor alpha-positive (PDGFRα(+)) mesenchymal cells from bone marrow (BM) into circulation. However, whether HMGB1-induced endogenous PDGFRα(+) mesenchymal cells stimulate skin regeneration has been unclear. Here, we investigated the functions of the HMGB1/BM-PDGFRα(+) mesenchymal cell axis in the regeneration of mouse skin grafts. We found that intravenous HMGB1 administration induced an accumulation of endogenous BM-PDGFRα(+) mesenchymal cells followed by significant inflammatory suppression in the grafts. In contrast, mice with reduced BM-PDGFRα(+) mesenchymal cells showed massive inflammation of the grafts compared to mice that had normal levels of these cells even after HMGB1 administration, suggesting that BM-PDGFRα(+) mesenchymal cells contribute to the HMGB1-induced anti-inflammatory effect. We also found that intravenously administered HMGB1 augmented the local migration of BM-PDGFRα(+) mesenchymal cells from circulation to skin graft by inducing the expression of CXCR4, an SDF-1 receptor, on these cells. Finally, we showed the therapeutic activity of the HMGB1/BM-PDGFRα(+) mesenchymal cell axis in an allergic contact dermatitis model. The results illustrated the contribution of the HMGB1/BM-PDGFRα(+) mesenchymal cell axis in suppressing the inflammation of injured/inflamed skin. These findings may provide future perspectives on the use of HMGB1-based medicines for intractable diseases.


Assuntos
Anti-Inflamatórios/administração & dosagem , Proteína HMGB1/administração & dosagem , Células-Tronco Mesenquimais/fisiologia , Animais , Células da Medula Óssea/fisiologia , Movimento Celular , Células Cultivadas , Dermatite Alérgica de Contato/tratamento farmacológico , Dermatite Alérgica de Contato/imunologia , Avaliação Pré-Clínica de Medicamentos , Masculino , Camundongos Endogâmicos C57BL , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Pele/efeitos dos fármacos , Pele/imunologia , Pele/patologia , Transplante de Pele , Regulação para Cima , Cicatrização
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA