Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 14(1): 8273, 2023 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-38092736

RESUMO

Adult tissue-resident macrophages (RMs) are either maintained by blood monocytes or through self-renewal. While the presence of a nurturing niche is likely crucial to support the survival and function of self-renewing RMs, evidence regarding its nature is limited. Here, we identify fibro-adipogenic progenitors (FAPs) as the main source of colony-stimulating factor 1 (CSF1) in resting skeletal muscle. Using parabiosis in combination with FAP-deficient transgenic mice (PdgfrαCreERT2 × DTA) or mice lacking FAP-derived CSF1 (PdgfrαCreERT2 × Csf1flox/null), we show that local CSF1 from FAPs is required for the survival of both TIM4- monocyte-derived and TIM4+ self-renewing RMs in adult skeletal muscle. The spatial distribution and number of TIM4+ RMs coincide with those of dipeptidyl peptidase IV (DPPIV)+ FAPs, suggesting their role as CSF1-producing niche cells for self-renewing RMs. This finding identifies opportunities to precisely manipulate the function of self-renewing RMs in situ to further unravel their role in health and disease.


Assuntos
Dipeptidil Peptidase 4 , Receptor alfa de Fator de Crescimento Derivado de Plaquetas , Camundongos , Animais , Diferenciação Celular/fisiologia , Dipeptidil Peptidase 4/genética , Adipogenia , Músculo Esquelético , Camundongos Transgênicos , Macrófagos
2.
Dev Cell ; 58(6): 489-505.e7, 2023 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-36898377

RESUMO

Loss of muscle mass is a common manifestation of chronic disease. We find the canonical Wnt pathway to be activated in mesenchymal progenitors (MPs) from cancer-induced cachectic mouse muscle. Next, we induce ß-catenin transcriptional activity in murine MPs. As a result, we observe expansion of MPs in the absence of tissue damage, as well as rapid loss of muscle mass. Because MPs are present throughout the organism, we use spatially restricted CRE activation and show that the induction of tissue-resident MP activation is sufficient to induce muscle atrophy. We further identify increased expression of stromal NOGGIN and ACTIVIN-A as key drivers of atrophic processes in myofibers, and we verify their expression by MPs in cachectic muscle. Finally, we show that blocking ACTIVIN-A rescues the mass loss phenotype triggered by ß-catenin activation in MPs, confirming its key functional role and strengthening the rationale for targeting this pathway in chronic disease.


Assuntos
Via de Sinalização Wnt , beta Catenina , Camundongos , Animais , beta Catenina/metabolismo , Ativinas , Músculos/metabolismo
3.
Proc Natl Acad Sci U S A ; 119(44): e2209976119, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36279473

RESUMO

IFNγ is traditionally known as a proinflammatory cytokine with diverse roles in antimicrobial and antitumor immunity. Yet, findings regarding its sources and functions during the regeneration process following a sterile injury are conflicting. Here, we show that natural killer (NK) cells are the main source of IFNγ in regenerating muscle. Beyond this cell population, IFNγ production is limited to a small population of T cells. We further show that NK cells do not play a major role in muscle regeneration following an acute injury or in dystrophic mice. Surprisingly, the absence of IFNγ per se also has no effect on muscle regeneration following an acute injury. However, the role of IFNγ is partially unmasked when TNFα is also neutralized, suggesting a compensatory mechanism. Using transgenic mice, we showed that conditional inhibition of IFNGR1 signaling in muscle stem cells or fibro-adipogenic progenitors does not play a major role in muscle regeneration. In contrast to common belief, we found that IFNγ is not present in the early inflammatory phase of the regeneration process but rather peaks when macrophages are acquiring an anti-inflammatory phenotype. Further transcriptomic analysis suggests that IFNγ cooperates with TNFα to regulate the transition of macrophages from pro- to anti-inflammatory states. The absence of the cooperative effect of these cytokines on macrophages, however, does not result in significant regeneration impairment likely due to the presence of other compensatory mechanisms. Our findings support the arising view of IFNγ as a pleiotropic inflammatory regulator rather than an inducer of the inflammatory response.


Assuntos
Macrófagos , Fator de Necrose Tumoral alfa , Camundongos , Animais , Interferon gama , Citocinas , Regeneração , Anti-Inflamatórios , Músculos
4.
Sci Transl Med ; 14(651): eabg7504, 2022 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-35767650

RESUMO

The role of tissue-resident macrophages during tissue regeneration or fibrosis is not well understood, mainly due to the lack of a specific marker for their identification. Here, we identified three populations of skeletal muscle-resident myelomonocytic cells: a population of macrophages positive for lymphatic vessel endothelial receptor 1 (LYVE1) and T cell membrane protein 4 (TIM4 or TIMD4), a population of LYVE1-TIM4- macrophages, and a population of cells likely representing dendritic cells that were positive for CD11C and major histocompatibility complex class II (MHCII). Using a combination of parabiosis and lineage-tracing experiments, we found that, at steady state, TIM4- macrophages were replenished from the blood, whereas TIM4+ macrophages locally self-renewed [self-renewing resident macrophages (SRRMs)]. We further showed that Timd4 could be reliably used to distinguish SRRMs from damage-induced infiltrating macrophages. Using a colony-stimulating factor 1 receptor (CSF1R) inhibition/withdrawal approach to specifically deplete SRRMs, we found that SRRMs provided a nonredundant function in clearing damage-induced apoptotic cells early after extensive acute injury. In contrast, in chronic mild injury as seen in a mouse model of Duchenne muscular dystrophy, depletion of both TIM4-- and TIM4+-resident macrophage populations through long-term CSF1R inhibition changed muscle fiber composition from damage-sensitive glycolytic fibers toward damage-resistant glycolytic-oxidative fibers, thereby protecting muscle against contraction-induced injury both ex vivo and in vivo. This work reveals a previously unidentified role for resident macrophages in modulating tissue metabolism and may have therapeutic potential given the ongoing clinical testing of CSF1R inhibitors.


Assuntos
Macrófagos , Músculo Esquelético , Distrofias Musculares , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos , Animais , Macrófagos/metabolismo , Macrófagos/patologia , Proteínas de Membrana/metabolismo , Camundongos , Monócitos/metabolismo , Monócitos/patologia , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Distrofias Musculares/tratamento farmacológico , Distrofias Musculares/metabolismo , Distrofias Musculares/patologia , Receptores Proteína Tirosina Quinases/metabolismo , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/antagonistas & inibidores , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/metabolismo , Proteínas de Transporte Vesicular/metabolismo
5.
Exp Neurol ; 334: 113436, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32814068

RESUMO

Despite the ability of peripheral nerves to regenerate after injury, failure occurs due to an inability of supporting cells to maintain growth, resulting in long-term consequences such as sensorimotor dysfunction and neuropathic pain. Here, we investigate the potential of engaging the cellular adaptive response to hypoxia, via inhibiting its negative regulators, to enhance the regenerative process. Under normoxic conditions, prolyl hydroxylase domain (PHD) proteins 1, 2, and 3 hydroxylate the key metabolic regulator hypoxia inducible factor 1α (HIF1α), marking it for subsequent proteasomal degradation. We inhibited PHD protein function systemically via either individual genetic deletion or pharmacological pan-PHD inhibition using dimethyloxalylglycine (DMOG). We show enhanced axonal regeneration after sciatic nerve crush injury in PHD1-/- mice, PHD3-/- mice, and in DMOG-treated mice, and in PHD1-/- and DMOG-treated mice a reduction in hypersensitivity to cooling after permanent sciatic ligation. Electromyographically, PHD1-/- and PHD3-/- mice showed an increased CMAP amplitude one-month post-injury, probably due to protection against denervation induced muscle atrophy, while DMOG-treated and PHD2+/- mice showed reduced latencies, indicating improved motor axon function. DMOG treatment did not affect the growth of dorsal root ganglion neurites in vitro, suggesting a lack of direct effects of DMOG on axonal regrowth. Enhanced regeneration in vivo was concurrent with an increase in macrophage density, and a shift in macrophage polarization state ratios (from M1-like toward M2-like) in DMOG-treated animals. These results indicate PHD proteins as a novel therapeutic target to improve regenerative and functional outcomes after peripheral nerve injury without manipulating molecular O2.


Assuntos
Axônios/fisiologia , Hipóxia/metabolismo , Regeneração Nervosa/fisiologia , Traumatismos dos Nervos Periféricos/metabolismo , Recuperação de Função Fisiológica/fisiologia , Aminoácidos Dicarboxílicos/farmacologia , Aminoácidos Dicarboxílicos/uso terapêutico , Animais , Axônios/efeitos dos fármacos , Células Cultivadas , Hipóxia/tratamento farmacológico , Hipóxia/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Regeneração Nervosa/efeitos dos fármacos , Traumatismos dos Nervos Periféricos/tratamento farmacológico , Traumatismos dos Nervos Periféricos/patologia , Prolil Hidroxilases/metabolismo , Inibidores de Prolil-Hidrolase/farmacologia , Inibidores de Prolil-Hidrolase/uso terapêutico , Recuperação de Função Fisiológica/efeitos dos fármacos
6.
Skelet Muscle ; 6: 34, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27757223

RESUMO

BACKGROUND: The skeletal muscle stem cell niche provides an environment that maintains quiescent satellite cells, required for skeletal muscle homeostasis and regeneration. Syndecan-3, a transmembrane proteoglycan expressed in satellite cells, supports communication with the niche, providing cell interactions and signals to maintain quiescent satellite cells. RESULTS: Syndecan-3 ablation unexpectedly improves regeneration in repeatedly injured muscle and in dystrophic mice, accompanied by the persistence of sublaminar and interstitial, proliferating myoblasts. Additionally, muscle aging is improved in syndecan-3 null mice. Since syndecan-3 null myofiber-associated satellite cells downregulate Pax7 and migrate away from the niche more readily than wild type cells, syxndecan-3 appears to regulate satellite cell homeostasis and satellite cell homing to the niche. CONCLUSIONS: Manipulating syndecan-3 provides a promising target for development of therapies to enhance muscle regeneration in muscular dystrophies and in aged muscle.


Assuntos
Homeostase , Músculo Esquelético/fisiologia , Regeneração , Células Satélites de Músculo Esquelético/fisiologia , Nicho de Células-Tronco , Sindecana-3/fisiologia , Animais , Feminino , Masculino , Camundongos , Camundongos Knockout , Músculo Esquelético/lesões , Músculo Esquelético/patologia , Células Satélites de Músculo Esquelético/patologia , Sindecana-3/genética
7.
Nat Med ; 21(7): 786-94, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26053624

RESUMO

Depending on the inflammatory milieu, injury can result either in a tissue's complete regeneration or in its degeneration and fibrosis, the latter of which could potentially lead to permanent organ failure. Yet how inflammatory cells regulate matrix-producing cells involved in the reparative process is unknown. Here we show that in acutely damaged skeletal muscle, sequential interactions between multipotent mesenchymal progenitors and infiltrating inflammatory cells determine the outcome of the reparative process. We found that infiltrating inflammatory macrophages, through their expression of tumor necrosis factor (TNF), directly induce apoptosis of fibro/adipogenic progenitors (FAPs). In states of chronic damage, however, such as those in mdx mice, macrophages express high levels of transforming growth factor ß1 (TGF-ß1), which prevents the apoptosis of FAPs and induces their differentiation into matrix-producing cells. Treatment with nilotinib, a kinase inhibitor with proposed anti-fibrotic activity, can block the effect of TGF-ß1 and reduce muscle fibrosis in mdx mice. Our findings reveal an unexpected anti-fibrotic role of TNF and suggest that disruption of the precisely timed progression from a TNF-rich to a TGF-ß-rich environment favors fibrotic degeneration of the muscle during chronic injury.


Assuntos
Adipogenia/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Músculo Esquelético/lesões , Doenças Musculares/tratamento farmacológico , Pirimidinas/uso terapêutico , Células-Tronco/citologia , Fator de Necrose Tumoral alfa/farmacologia , Animais , Contagem de Células , Sobrevivência Celular/efeitos dos fármacos , Doença Crônica , Colágeno/metabolismo , Venenos Elapídicos , Feminino , Fibrose , Citometria de Fluxo , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Monócitos/citologia , Monócitos/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/patologia , Doenças Musculares/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores CCR2/deficiência , Receptores CCR2/metabolismo , Regeneração/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta1/metabolismo
8.
Cytotherapy ; 14(8): 917-24, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22687188

RESUMO

BACKGROUND AIMS: The clinical application of mesenchymal stromal cells (MSC) faces several obstacles, such as the lack of a standard method for direct isolation as well as a low frequency and concern about the safety of their in vitro expansion. Although the density-gradient separation technique is used as the first step in most methods of MSC isolation to enrich mononuclear cells, the efficiency of this method has not so far been examined. This study was designed to address this issue. METHODS: Human bone marrow (BM) samples were laid over Ficoll-Paque, and after centrifugation the upper and lower fractions were cultured separately. Surface markers, differentiation potential and the number of emerged cells were determined. RESULTS: The isolated cells from both the upper and lower fractions were characteristic of MSC. Although it is commonly believed that MSC are single suspending mononuclear cells and so are enriched in the upper fraction of Ficoll-Paque after density-gradient separation, our data showed that considerable numbers of these cells were accumulated in the lower fraction. Further data indicated that MSC were actually present as cell aggregates in BM and they could be enriched effectively by a simple filtration method. CONCLUSIONS: The aggregate nature of MSC in BM is in agreement with the concept that they are one of the main elements of the hematopoietic stem cell niche. In addition, the simple filtration method proposed here to isolate cell aggregates may provide opportunities for instant stem cell therapy without the need for extensive in vitro expansion.


Assuntos
Células da Medula Óssea/citologia , Técnicas de Cultura de Células , Células-Tronco Mesenquimais/citologia , Diferenciação Celular , Meios de Cultura , Humanos , Leucócitos Mononucleares/citologia
10.
Tissue Eng Part A ; 17(9-10): 1209-18, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21143044

RESUMO

Application of nanofibers for the purpose of tissue mimicking and regeneration has become widespread in the field of biomedicine. In this study, polyethersulfone (PES) electrospun nanofibrous membranes were fabricated, modified, and loaded with unrestricted somatic stem cells (USSC) to mimic the natural structure of bone. Untreated PES, plasma-treated PES, and collagen-grafted PES (COL-PES) nanofibers were characterized via Brunauer-Emmett-Teller method, attenuated total reflection Fourier transform infrared, contact angle measurements, and scanning electron microscopy. Their capacity to support proliferation, infiltration, and osteogenic differentiation of USSC was investigated using MTT assay, real-time reverse transcriptase-polymerase chain reaction, histologic staining, alkaline phosphatase activity, and calcium content assay. All the scaffolds had nanofibrous and highly porous structure with large surface area. After surface treatments, hydrophilicity of scaffolds increased intensively and their biocompatibility improved. During osteogenic differentiation of stem cells, alkaline phosphatase activity and calcium content exhibited the highest level in cells on COL-PES. Real-time reverse transcriptase-polymerase chain reaction showed significant difference between the expression levels of osteoblast-related genes on COL-PES compared to other scaffolds. Excellent infiltration of USSC was observed in nanofibrous membranes especially COL-PES. It can be concluded that COL-PES nanofibrous scaffold has potential for bone grafting because of its three-dimensional structure and bioactivity which enhance proliferation, differentiation, and infiltration of USSC.


Assuntos
Calcificação Fisiológica , Colágeno/química , Membranas Artificiais , Nanofibras/química , Osteoblastos/metabolismo , Células-Tronco/metabolismo , Antígenos de Diferenciação/biossíntese , Diferenciação Celular , Movimento Celular , Proliferação de Células , Células Cultivadas , Humanos , Osteoblastos/citologia , Polímeros/química , Porosidade , Células-Tronco/citologia , Sulfonas/química
11.
Tissue Eng Part A ; 16(11): 3527-36, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20624004

RESUMO

INTRODUCTION: In this study, thick polyethersulfone (PES) nanofibrous scaffolds were prepared by fine tuning of electrospinning parameters and was evaluated for wound dressing applications. MATERIALS AND METHODS: Scanning electron microscopy and Brunauer-Emmett-Teller methods were used for PES nanofibers characterization. The interaction between fibroblasts and nanofibers was studied in vitro. Further, a mouse model was used to evaluate the effectiveness of the PES scaffold in wound healing. Vaseline gauze dressing and a conventional gas permeable bandage were used as a control. The wound repair process was evaluated by histological examination and immunohistochemistry staining using antibodies to cytokeratin 10 (CK10), proliferating cell nuclear antigen (PCNA), and alpha-smooth muscle actin (alpha-SMA). RESULTS AND CONCLUSION: The characterization of nanofibers showed that the PES membrane has nanoscale, porous, high surface area structure. These properties conferred higher exudate absorption capacity for the PES scaffold which is essential for effective wound healing. In vitro results indicated that the PES scaffold can support fibroblast proliferation similar to that with tissue culture polystyrene. Epithelial regeneration was expeditiously accelerated under PES as compared with Vaseline gauze. Greater fibroblast maturation, improved collagen deposition and faster edema resolution were the superior properties of PES over the commercial dressing. Based on these results we conclude that the biocompatible PES nanofibers can effectively be used as a dressing to accelerate wound healing.


Assuntos
Derme/fisiologia , Epiderme/fisiologia , Nanofibras/química , Procedimentos de Cirurgia Plástica/métodos , Polímeros/farmacologia , Regeneração/efeitos dos fármacos , Sulfonas/farmacologia , Animais , Materiais Biocompatíveis/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Derme/efeitos dos fármacos , Epiderme/efeitos dos fármacos , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/ultraestrutura , Queratina-10/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Nanofibras/ultraestrutura , Polímeros/química , Antígeno Nuclear de Célula em Proliferação/metabolismo , Coloração e Rotulagem , Sulfonas/química , Cicatrização/efeitos dos fármacos
12.
Biochem Biophys Res Commun ; 382(1): 129-33, 2009 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-19265673

RESUMO

Nanofibrous scaffolds have been recently used in the field of tissue engineering because of their nano-size structure which promotes cell attachment, function, proliferation and infiltration. In this study, nanofibrous polyethersulfone (PES) scaffolds was prepared via electrospinning. The scaffolds were surface modified by plasma treatment and collagen grafting. The surface changes then investigated by contact angle measurements and FTIR-ATR. The results proved grafting of the collagen on nanofibers surface and increased hydrophilicity after plasma treatment and collagen grafting. The cell interaction study was done using stem cells because of their ability to differentiate to different kinds of cell lines. The cells had normal morphology on nanofibers and showed very high infiltration through collagen grafted PES nanofibers. This infiltration capability is very useful and needed to make 3D scaffolds in tissue engineering.


Assuntos
Nanoestruturas/química , Polímeros/química , Células-Tronco/fisiologia , Sulfonas/química , Engenharia Tecidual , Alicerces Teciduais/química , Células Imobilizadas , Humanos , Células-Tronco/química , Células-Tronco/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...