Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
FASEB J ; 34(10): 13521-13532, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32794246

RESUMEN

Inflammaging is associated with poor tissue regeneration observed in advanced age. Specifically, protracted inflammation after acute injury has been associated with decreased bone fracture healing and increased rates of nonunion in elderly patients. Here, we investigated the efficacy of using Maresin 1 (MaR1), an omega-3 fatty acid-derived pro-resolving agent, to resolve inflammation after tibial fracture injury and subsequently improving aged bone healing. Aged (24-month-old mice) underwent tibial fracture surgery and were either treated with vehicle or MaR1 3 days after injury. Fracture calluses were harvested 7 days, 14 days, 21 days, and 28 days after injury to investigate inflammatory response, cartilage development, bone deposition, and mechanical integrity, respectively. Healing bones from MaR1-treated mice displayed decreased cartilage formation and increased bone deposition which resulted in increased structural stiffness and increased force to fracture in the later stages of repair. In the early stages, MaR1 treatment decreased the number of pro-inflammatory macrophages within the fracture callus and decreased the level of inflammatory biomarkers in circulation. In tissue culture models, MaR1 treatment of bone marrow-derived macrophages from aged mice protected cells form a pro-inflammatory phenotype and induced an anti-inflammatory fate. Furthermore, the secretome of MaR1-treated bone marrow-derived macrophages was identified as osteoinductive, enhancing osteoblast differentiation of bone marrow stromal cells. Our findings here identify resolution of inflammation, and MaR1 itself, to be a point of intervention to improve aged bone healing.


Asunto(s)
Antiinflamatorios , Regeneración Ósea/efectos de los fármacos , Ácidos Docosahexaenoicos , Curación de Fractura/efectos de los fármacos , Inflamación/tratamiento farmacológico , Fracturas de la Tibia/tratamiento farmacológico , Envejecimiento , Animales , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Ácidos Docosahexaenoicos/farmacología , Ácidos Docosahexaenoicos/uso terapéutico , Femenino , Macrófagos/citología , Masculino , Células Madre Mesenquimatosas/citología , Ratones
2.
J Neuroinflammation ; 16(1): 193, 2019 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-31660984

RESUMEN

BACKGROUND: Patients with pre-existing neurodegenerative disease commonly experience fractures that require orthopedic surgery. Perioperative neurocognitive disorders (PND), including delirium and postoperative cognitive dysfunction, are serious complications that can result in increased 1-year mortality when superimposed on dementia. Importantly, there are no disease-modifying therapeutic options for PND. Our lab developed the "broad spectrum" mixed-lineage kinase 3 inhibitor URMC-099 to inhibit pathological innate immune responses that underlie neuroinflammation-associated cognitive dysfunction. Here, we test the hypothesis that URMC-099 can prevent surgery-induced neuroinflammation and cognitive impairment. METHODS: Orthopedic surgery was performed by fracturing the tibia of the left hindlimb with intramedullary fixation under general anesthesia and analgesia. In a pilot experiment, 9-month-old mice were treated five times with URMC-099 (10 mg/kg, i.p.), spaced 12 h apart, with three doses prior to surgery and two doses following surgery. In this experiment, microgliosis was evaluated using unbiased stereology and blood-brain barrier (BBB) permeability was assessed using immunoglobulin G (IgG) immunostaining. In follow-up experiments, 3-month-old mice were treated only three times with URMC-099 (10 mg/kg, i.p.), spaced 12 h apart, prior to orthopedic surgery. Two-photon scanning laser microscopy and CLARITY with light-sheet microscopy were used to define surgery-induced changes in microglial dynamics and morphology, respectively. Surgery-induced memory impairment was assessed using the "What-Where-When" and Memory Load Object Discrimination tasks. The acute peripheral immune response to surgery was assessed by cytokine/chemokine profiling and flow cytometry. Finally, long-term fracture healing was assessed in fracture callouses using micro-computerized tomography (microCT) and histomorphometry analyses. RESULTS: Orthopedic surgery induced BBB disruption and microglial activation, but had no effect on microglial process motility. Surgically treated mice exhibited impaired object place and identity discrimination in the "What-Where-When" and Memory Load Object Discrimination tasks. Both URMC-099 dosing paradigms prevented the neuroinflammatory sequelae that accompanied orthopedic surgery. URMC-099 prophylaxis had no effect on the mobilization of the peripheral innate immune response and fracture healing. CONCLUSIONS: These findings show that prophylactic URMC-099 treatment is sufficient to prevent surgery-induced microgliosis and cognitive impairment without affecting fracture healing. Together, these findings provide compelling evidence for the advancement of URMC-099 as a therapeutic option for PND.


Asunto(s)
Disfunción Cognitiva/prevención & control , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Microglía/efectos de los fármacos , Atención Perioperativa , Piridinas/uso terapéutico , Pirroles/uso terapéutico , Animales , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/patología , Modelos Animales de Enfermedad , Femenino , Quinasas Quinasa Quinasa PAM/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/metabolismo , Microglía/patología , Trastornos Neurocognitivos/tratamiento farmacológico , Trastornos Neurocognitivos/metabolismo , Trastornos Neurocognitivos/patología , Atención Perioperativa/métodos , Piridinas/farmacología , Pirroles/farmacología , Proteina Quinasa Quinasa Quinasa 11 Activada por Mitógeno
3.
Curr Osteoporos Rep ; 16(2): 138-145, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29508143

RESUMEN

PURPOSE OF REVIEW: Bone fracture healing is a complex physiological process relying on numerous cell types and signals. Inflammatory factors secreted by immune cells help to control recruitment, proliferation, differentiation, and activation of hematopoietic and mesenchymal cells. Within this review we will discuss the functional role of immune cells as it pertains to bone fracture healing. In doing so, we will outline the cytokines secreted and their effects within the healing fracture callus. RECENT FINDINGS: Macrophages have been found to play an important role in fracture healing. These immune cells signal to other cells of the fracture callus, modulating bone healing. Cytokines and cellular signals within fracture healing continue to be studied. The findings from this work have helped to reinforce the importance of osteoimmunity in bone fracture healing. Owing to these efforts, immunomodulation is emerging as a potential therapeutic target to improve bone fracture healing.


Asunto(s)
Callo Óseo/inmunología , Citocinas/inmunología , Curación de Fractura/inmunología , Macrófagos/inmunología , Diferenciación Celular/inmunología , Proliferación Celular , Células Madre Hematopoyéticas , Humanos , Células Madre Mesenquimatosas
4.
Development ; 140(12): 2597-610, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23715552

RESUMEN

Osteoarthritis primarily affects the articular cartilage of synovial joints. Cell and/or cartilage replacement is a promising therapy, provided there is access to appropriate tissue and sufficient numbers of articular chondrocytes. Embryonic stem cells (ESCs) represent a potentially unlimited source of chondrocytes and tissues as they can generate a broad spectrum of cell types under appropriate conditions in vitro. Here, we demonstrate that mouse ESC-derived chondrogenic mesoderm arises from a Flk-1(-)/Pdgfrα(+) (F(-)P(+)) population that emerges in a defined temporal pattern following the development of an early cardiogenic F(-)P(+) population. Specification of the late-arising F(-)P(+) population with BMP4 generated a highly enriched population of chondrocytes expressing genes associated with growth plate hypertrophic chondrocytes. By contrast, specification with Gdf5, together with inhibition of hedgehog and BMP signaling pathways, generated a population of non-hypertrophic chondrocytes that displayed properties of articular chondrocytes. The two chondrocyte populations retained their hypertrophic and non-hypertrophic properties when induced to generate spatially organized proteoglycan-rich cartilage-like tissue in vitro. Transplantation of either type of chondrocyte, or tissue generated from them, into immunodeficient recipients resulted in the development of cartilage tissue and bone within an 8-week period. Significant ossification was not observed when the tissue was transplanted into osteoblast-depleted mice or into diffusion chambers that prevent vascularization. Thus, through stage-specific manipulation of appropriate signaling pathways it is possible to efficiently and reproducibly derive hypertrophic and non-hypertrophic chondrocyte populations from mouse ESCs that are able to generate distinct cartilage-like tissue in vitro and maintain a cartilage tissue phenotype within an avascular and/or osteoblast-free niche in vivo.


Asunto(s)
Cartílago Articular/citología , Condrocitos/citología , Condrogénesis , Células Madre Embrionarias/citología , Animales , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/metabolismo , Cartílago Articular/metabolismo , Diferenciación Celular , Linaje de la Célula , Condrocitos/metabolismo , Células Madre Embrionarias/metabolismo , Células Madre Embrionarias/trasplante , Femenino , Factor 5 de Diferenciación de Crecimiento/genética , Factor 5 de Diferenciación de Crecimiento/metabolismo , Hipertrofia/metabolismo , Inmunohistoquímica , Mesodermo/citología , Mesodermo/metabolismo , Ratones , Osteoblastos/metabolismo , Osteogénesis , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal , Factores de Tiempo
5.
Calcif Tissue Int ; 97(5): 476-86, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26220824

RESUMEN

Age-related bone loss may be a result of declining levels of stem cells in the bone marrow. Using the Col2.3Δtk (DTK) transgenic mouse, osteoblast depletion was used as a source of marrow stress in order to investigate the effects of aging on osteogenic progenitors which reside in the marrow space. Five-month-old DTK mice were treated with one or two cycles of ganciclovir to conditionally ablate differentiated osteoblasts, whereas controls were saline-treated. Treatment cycles were two weeks in length followed by four weeks of recovery. All animals were sacrificed at 8 months of age; bone marrow stromal cells (BMSCs) were harvested for cell culture and whole bones were excised for bone quality assessment. Colony-forming unit (CFU) assays were conducted to investigate the osteogenic potential of BMSC in vitro, and RNA was extracted to assess the expression of osteoblastic genes. Bone quality assessments included bone histomorphometry, TRAP staining, microcomputed tomography, and biomechanical testing. Osteoblast depletion decreased CFU-F (fibroblast), CFU-ALP (alkaline phosphatase), and CFU-VK (von Kossa) counts and BMSC osteogenic capacity in cell culture. Ex vivo, there were no differences in bone mineral density of vertebrae or femurs between treatment groups. Histology showed a decrease in bone volume and bone connectivity with repeated osteoblast depletion; however, this was accompanied by an increase in bone formation rate. There were no notable differences in osteoclast parameters or observed bone marrow adiposity. We have developed a model that uses bone marrow stress to mimic age-related decrease in osteogenic progenitors. Our data suggest that the number of healthy BMSCs and their osteogenic potential decline with repeated osteoblast depletion. However, activity of the remaining osteoblasts increases to compensate for this loss in progenitor osteogenic potential.


Asunto(s)
Células Madre Mesenquimatosas/metabolismo , Osteogénesis/fisiología , Células Madre/metabolismo , Estrés Fisiológico , Envejecimiento , Animales , Fenómenos Biomecánicos , Densidad Ósea , Huesos/diagnóstico por imagen , Modelos Animales de Enfermedad , Células Madre Mesenquimatosas/patología , Ratones , Ratones Transgénicos , Osteoblastos/metabolismo , Osteoblastos/patología , Reacción en Cadena en Tiempo Real de la Polimerasa , Células Madre/patología , Microtomografía por Rayos X
6.
Cell Rep ; 43(3): 113881, 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38442019

RESUMEN

An intriguing effect of short-term caloric restriction (CR) is the expansion of certain stem cell populations, including muscle stem cells (satellite cells), which facilitate an accelerated regenerative program after injury. Here, we utilized the MetRSL274G (MetRS) transgenic mouse to identify liver-secreted plasminogen as a candidate for regulating satellite cell expansion during short-term CR. Knockdown of circulating plasminogen prevents satellite cell expansion during short-term CR. Furthermore, loss of the plasminogen receptor KT (Plg-RKT) is also sufficient to prevent CR-related satellite cell expansion, consistent with direct signaling of plasminogen through the plasminogen receptor Plg-RKT/ERK kinase to promote proliferation of satellite cells. Importantly, we are able to replicate many of these findings in human participants from the CALERIE trial. Our results demonstrate that CR enhances liver protein secretion of plasminogen, which signals directly to the muscle satellite cell through Plg-RKT to promote proliferation and subsequent muscle resilience during CR.


Asunto(s)
Plasminógeno , Receptores de Superficie Celular , Ratones , Animales , Humanos , Plasminógeno/metabolismo , Receptores de Superficie Celular/metabolismo , Restricción Calórica , Hígado/metabolismo , Ratones Transgénicos , Serina Proteasas , Proliferación Celular , Músculos/metabolismo
7.
Nat Aging ; 3(8): 948-964, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37500973

RESUMEN

Heterochronic parabiosis (HPB) is known for its functional rejuvenation effects across several mouse tissues. However, its impact on biological age and long-term health is unknown. Here we performed extended (3-month) HPB, followed by a 2-month detachment period of anastomosed pairs. Old detached mice exhibited improved physiological parameters and lived longer than control isochronic mice. HPB drastically reduced the epigenetic age of blood and liver based on several clock models using two independent platforms. Remarkably, this rejuvenation effect persisted even after 2 months of detachment. Transcriptomic and epigenomic profiles of anastomosed mice showed an intermediate phenotype between old and young, suggesting a global multi-omic rejuvenation effect. In addition, old HPB mice showed gene expression changes opposite to aging but akin to several life span-extending interventions. Altogether, we reveal that long-term HPB results in lasting epigenetic and transcriptome remodeling, culminating in the extension of life span and health span.


Asunto(s)
Longevidad , Rejuvenecimiento , Ratones , Animales , Longevidad/genética , Multiómica , Envejecimiento/genética
8.
Cell Metab ; 35(5): 807-820.e5, 2023 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-37086720

RESUMEN

Aging is classically conceptualized as an ever-increasing trajectory of damage accumulation and loss of function, leading to increases in morbidity and mortality. However, recent in vitro studies have raised the possibility of age reversal. Here, we report that biological age is fluid and exhibits rapid changes in both directions. At epigenetic, transcriptomic, and metabolomic levels, we find that the biological age of young mice is increased by heterochronic parabiosis and restored following surgical detachment. We also identify transient changes in biological age during major surgery, pregnancy, and severe COVID-19 in humans and/or mice. Together, these data show that biological age undergoes a rapid increase in response to diverse forms of stress, which is reversed following recovery from stress. Our study uncovers a new layer of aging dynamics that should be considered in future studies. The elevation of biological age by stress may be a quantifiable and actionable target for future interventions.


Asunto(s)
COVID-19 , Humanos , Animales , Ratones , Envejecimiento/fisiología , Parabiosis
9.
Cell Stem Cell ; 30(1): 96-111.e6, 2023 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-36516837

RESUMEN

The efficacy and safety of gene-therapy strategies for indications like tissue damage hinge on precision; yet, current methods afford little spatial or temporal control of payload delivery. Here, we find that tissue-regeneration enhancer elements (TREEs) isolated from zebrafish can direct targeted, injury-associated gene expression from viral DNA vectors delivered systemically in small and large adult mammalian species. When employed in combination with CRISPR-based epigenome editing tools in mice, zebrafish TREEs stimulated or repressed the expression of endogenous genes after ischemic myocardial infarction. Intravenously delivered recombinant AAV vectors designed with a TREE to direct a constitutively active YAP factor boosted indicators of cardiac regeneration in mice and improved the function of the injured heart. Our findings establish the application of contextual enhancer elements as a potential therapeutic platform for spatiotemporally controlled tissue regeneration in mammals.


Asunto(s)
Elementos de Facilitación Genéticos , Terapia Genética , Corazón , Infarto del Miocardio , Miocitos Cardíacos , Regeneración , Animales , Ratones , Proliferación Celular , Corazón/fisiología , Infarto del Miocardio/genética , Infarto del Miocardio/terapia , Miocitos Cardíacos/metabolismo , Pez Cebra/genética , Terapia Genética/métodos , Regeneración/genética
10.
Nat Commun ; 13(1): 7613, 2022 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-36494364

RESUMEN

Pathologies associated with sarcopenia include decline in muscular strength, lean mass and regenerative capacity. Despite the substantial impact on quality of life, no pharmacological therapeutics are available to counteract the age-associated decline in functional capacity and/or, resilience. Evidence suggests immune-secreted cytokines can improve muscle regeneration, a strategy which we leverage in this study by rescuing the age-related deficiency in Meteorin-like through several in vivo add-back models. Notably, the intramuscular, peptide injection of recombinant METRNL was sufficient to improve muscle regeneration in aging. Using ex vivo media exchange and in vivo TNF inhibition, we demonstrate a mechanism of METRNL action during regeneration, showing it counteracts a pro-fibrotic gene program by triggering TNFα-induced apoptosis of fibro/adipogenic progenitor cells. These findings demonstrate therapeutic applications for METRNL to improve aged muscle, and show Fibro/Adipogenic Progenitors are viable therapeutic targets to counteract age-related loss in muscle resilience.


Asunto(s)
Músculo Esquelético , Calidad de Vida , Músculo Esquelético/fisiología , Adipogénesis , Células Madre , Citocinas
11.
J Orthop Res ; 40(11): 2510-2521, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35076116

RESUMEN

Meteorin-like protein (Metrnl), homologous to the initially identified neurotrophic factor Meteorin, is a secreted, multifunctional protein. Here we used mouse models to investigate Metrnl's role in skeletal development and bone fracture healing. During development Metrnl was expressed in the perichondrium and primary ossification center. In neonates, single cell RNA-seq of diaphyseal bone demonstrated strongest expression of Metrnl transcript by osteoblasts. In vitro, Metrnl was osteoinductive, increasing osteoblast differentiation and mineralization in tissue culture models. In vivo, loss of Metrnl expression resulted in no change in skeletal metrics in utero, at birth, or during postnatal growth. Six-week-old Metrnl-null mice displayed similar body length, body weight, tibial length, femoral length, BV/TV, trabecular number, trabecular thickness, and cortical thickness as littermate controls. In 4-month-old mice, lack of Metrnl expression did not change structural stiffness, ultimate force, or energy to fracture of femora under 3-point-bending. Last, we investigated the role of Metrnl in bone fracture healing. Metrnl expression increased in response to tibial injury, however, loss of Metrnl expression did not affect the amount of bone deposited within the healing tissue nor did it change the structural parameters of healing tissue. This work identifies Metrnl as a dispensable molecule for skeletal development. However, the osteoinductive capabilities of Metrnl may be utilized to modulate osteoblast differentiation in cell-based orthopedic therapies.


Asunto(s)
Curación de Fractura , Factores de Crecimiento Nervioso , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factores de Crecimiento Nervioso/metabolismo , Osteoblastos/metabolismo
12.
Biochem J ; 428(3): 385-95, 2010 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-20377527

RESUMEN

Acidic phosphoproteins of mineralized tissues such as bone and dentin are believed to play important roles in HA (hydroxyapatite) nucleation and growth. BSP (bone sialoprotein) is the most potent known nucleator of HA, an activity that is thought to be dependent on phosphorylation of the protein. The present study identifies the role phosphate groups play in mineral formation. Recombinant BSP and peptides corresponding to residues 1-100 and 133-205 of the rat sequence were phosphorylated with CK2 (protein kinase CK2). Phosphorylation increased the nucleating activity of BSP and BSP-(133-205), but not BSP-(1-100). MS analysis revealed that the major site phosphorylated within BSP-(133-205) was Ser136, a site adjacent to the series of contiguous glutamate residues previously implicated in HA nucleation. The critical role of phosphorylated Ser136 in HA nucleation was confirmed by site-directed mutagenesis and functional analyses. Furthermore, peptides corresponding to the 133-148 sequence of rat BSP were synthesized with or without a phosphate group on Ser136. As expected, the phosphopeptide was a more potent nucleator. The mechanism of nucleation was investigated using molecular-dynamics simulations analysing BSP-(133-148) interacting with the {100} crystal face of HA. Both phosphorylated and non-phosphorylated sequences adsorbed to HA in extended conformations with alternating residues in contact with and facing away from the crystal face. However, this alternating-residue pattern was more pronounced when Ser136 was phosphorylated. These studies demonstrate a critical role for Ser136 phosphorylation in BSP-mediated HA nucleation and identify a unique mode of interaction between the nucleating site of the protein and the {100} face of HA.


Asunto(s)
Durapatita/química , Serina/metabolismo , Sialoglicoproteínas/metabolismo , Animales , Sitios de Unión , Durapatita/metabolismo , Sialoproteína de Unión a Integrina , Mutagénesis Sitio-Dirigida , Fosforilación , Ratas , Serina/genética , Sialoglicoproteínas/química
13.
Adv Mater ; 32(8): e1906022, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31833120

RESUMEN

Approaches that enable innate repair mechanisms hold great potential for tissue repair. Herein, biomaterial-assisted sequestration of small molecules is described to localize pro-regenerative signaling at the injury site. Specifically, a synthetic biomaterial containing boronate molecules is designed to sequester adenosine, a small molecule ubiquitously present in the human body. The biomaterial-assisted sequestration of adenosine leverages the transient surge of extracellular adenosine following injury to prolong local adenosine signaling. It is demonstrated that implantation of the biomaterial patch following injury establishes an in situ stockpile of adenosine, resulting in accelerated healing by promoting both osteoblastogenesis and angiogenesis. The adenosine content within the patch recedes to the physiological level as the tissue regenerates. In addition to sequestering endogenous adenosine, the biomaterial is also able to deliver exogenous adenosine to the site of injury, offering a versatile solution to utilizing adenosine as a potential therapeutic for tissue repair.


Asunto(s)
Adenosina/química , Materiales Biocompatibles/química , Cicatrización de Heridas , Adenosina/metabolismo , Animales , Materiales Biocompatibles/farmacología , Materiales Biocompatibles/uso terapéutico , Diferenciación Celular , Fracturas Óseas/terapia , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Osteogénesis , Andamios del Tejido/química , Cicatrización de Heridas/efectos de los fármacos
14.
Nat Metab ; 2(3): 278-289, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32694780

RESUMEN

The immune system plays a multifunctional role throughout the regenerative process, regulating both pro-/anti-inflammatory phases and progenitor cell function. In the present study, we identify the myokine/cytokine Meteorin-like (Metrnl) as a critical regulator of muscle regeneration. Mice genetically lacking Metrnl have impaired muscle regeneration associated with a reduction in immune cell infiltration and an inability to transition towards an anti-inflammatory phenotype. Isochronic parabiosis, joining wild-type and whole-body Metrnl knock-out (KO) mice, returns Metrnl expression in the injured muscle and improves muscle repair, providing supportive evidence for Metrnl secretion from infiltrating immune cells. Macrophage-specific Metrnl KO mice are also deficient in muscle repair. During muscle regeneration, Metrnl works, in part, through Stat3 activation in macrophages, resulting in differentiation to an anti-inflammatory phenotype. With regard to myogenesis, Metrnl induces macrophage-dependent insulin-like growth factor 1 production, which has a direct effect on primary muscle satellite cell proliferation. Perturbations in this pathway inhibit efficacy of Metrnl in the regenerative process. Together, these studies identify Metrnl as an important regulator of muscle regeneration and a potential therapeutic target to enhance tissue repair.


Asunto(s)
Factor I del Crecimiento Similar a la Insulina/metabolismo , Músculo Esquelético/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética
15.
16.
JCI Insight ; 4(18)2019 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-31534056

RESUMEN

Age is a well-established risk factor for impaired bone fracture healing. Here, we identify a role for apolipoprotein E (ApoE) in age-associated impairment of bone fracture healing and osteoblast differentiation, and we investigate the mechanism by which ApoE alters these processes. We identified that, in both humans and mice, circulating ApoE levels increase with age. We assessed bone healing in WT and ApoE-/- mice after performing tibial fracture surgery: bone deposition was higher within fracture calluses from ApoE-/- mice. In vitro recombinant ApoE (rApoE) treatment of differentiating osteoblasts decreased cellular differentiation and matrix mineralization. Moreover, this rApoE treatment decreased osteoblast glycolytic activity while increasing lipid uptake and fatty acid oxidation. Using parabiosis models, we determined that circulating ApoE plays a strong inhibitory role in bone repair. Using an adeno-associated virus-based siRNA system, we decreased circulating ApoE levels in 24-month-old mice and demonstrated that, as a result, fracture calluses from these aged mice displayed enhanced bone deposition and mechanical strength. Our results demonstrate that circulating ApoE as an aging factor inhibits bone fracture healing by altering osteoblast metabolism, thereby identifying ApoE as a new therapeutic target for improving bone repair in the elderly.


Asunto(s)
Envejecimiento/sangre , Apolipoproteínas E/sangre , Apolipoproteínas E/genética , Curación de Fractura/fisiología , Osteoblastos/fisiología , Fracturas de la Tibia/fisiopatología , Adulto , Factores de Edad , Anciano , Anciano de 80 o más Años , Envejecimiento/fisiología , Animales , Apolipoproteínas E/antagonistas & inhibidores , Callo Óseo/diagnóstico por imagen , Callo Óseo/efectos de los fármacos , Callo Óseo/fisiopatología , Calcificación Fisiológica/efectos de los fármacos , Calcificación Fisiológica/genética , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Células Cultivadas , Estudios de Cohortes , Dependovirus/genética , Modelos Animales de Enfermedad , Femenino , Curación de Fractura/efectos de los fármacos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Humanos , Ratones , Ratones Noqueados para ApoE , Persona de Mediana Edad , Osteoblastos/efectos de los fármacos , Cultivo Primario de Células , ARN Interferente Pequeño/genética , Proteínas Recombinantes/administración & dosificación , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Fracturas de la Tibia/diagnóstico por imagen , Fracturas de la Tibia/cirugía , Microtomografía por Rayos X
17.
Matrix Biol ; 27(7): 600-8, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18620053

RESUMEN

In bone, hydroxyapatite (HA) crystals are deposited onto the type I collagen scaffold by a mechanism that has yet to be elucidated. Bone sialoprotein (BSP) is an acidic phosphoprotein that is expressed at high levels in mineralized tissues, capable of binding type I collagen, and nucleating HA. Both bone-extracted and recombinant BSP (rBSP) bind with equal affinity to collagen. The nature of the BSP-collagen interaction and its role in HA nucleation are not known. We have used a solid-phase binding assay and affinity chromatography to characterize the BSP-collagen interaction. rBSP-binding affinities of triple-helical and fibrillar type I collagen were similar (K(D) approximately 13 nM), while that of heat-denatured type I collagen was lower (K(D) approximately 44 nM), indicating the importance of triple-helical structure in binding BSP. Pepsin treatment of collagen had no effect on rBSP binding, demonstrating that the telopeptides of collagen are not involved. The majority of collagen-bound rBSP was eluted by acetonitrile, indicating that hydrophobic interactions are principally responsible for binding. Using an HA-nucleation assay, it was shown that rBSP is ten-fold more potent in reconstituted fibrillar collagen gels than in agarose gels. Nucleating potency of a non-collagen-binding, HA-nucleating peptide [rBSP(134-206)] showed no difference in the two gel systems. The work here shows that optimal binding of rBSP requires collagen to be in a native, triple-helical structure, does not require the telopeptides, and is stabilized by hydrophobic interactions. Upon binding to collagen, rBSP displays an increase in nucleation potency, implying a co-operative effect of BSP and collagen in mineral formation.


Asunto(s)
Colágeno/metabolismo , Hidroxiapatitas/química , Sialoglicoproteínas/metabolismo , Animales , Huesos/metabolismo , Calcio/química , Cromatografía/métodos , Colágeno/química , Geles , Sialoproteína de Unión a Integrina , Cinética , Modelos Biológicos , Fosfatos/química , Unión Proteica , Ratas , Proteínas Recombinantes/química , Sefarosa/química
19.
J Vis Exp ; (132)2018 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-29553500

RESUMEN

Surgery is commonly used to improve and maintain quality of life. Unfortunately, in vulnerable patients such as the elderly, complications may occur and significantly diminish the outcome. Indeed, after routine orthopedic surgery to repair a fracture, as many as 50% of elderly patients suffer from neurologic complications like delirium. Also, the capacity to heal and regenerate tissue after surgery decreases with age, and can impact the quality of fracture repair and even osseous integration of implants. Thus, a better understanding of mechanisms that drive these age-dependent changes could provide strategic targets to minimize risk for such complications and optimize outcomes. Here, we introduce a clinically relevant mouse model of tibial fracture. The postoperative changes in these mice mimic some of the cognitive impairments commonly observed after routine orthopedic surgery in humans. Briefly, an incision is performed in the right hind limb under strictly aseptic conditions. Muscles are disassociated, and a 0.38-mm stainless steel pin is inserted into the upper crest of the tibia, inside the intramedullary canal. Osteotomy is then performed, and the wound is stapled. We have used this model to investigate the effects of surgical trauma on postoperative neuroinflammation and behavioral changes. By applying this fracture model in combination with parabiosis, a surgical model in which 2 mice are anastomosed, we have studied cells and secreted factors that systemically rejuvenate organ function and tissue regeneration after injury. By following our step-by-step protocol, these models can be reproduced with high fidelity, and can be adapted to interrogate many biologic pathways that are altered by surgical trauma.


Asunto(s)
Disfunción Cognitiva/etiología , Regeneración Tisular Dirigida/métodos , Ortopedia/métodos , Anciano , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Complicaciones Posoperatorias
20.
Nat Commun ; 9(1): 5191, 2018 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-30518764

RESUMEN

The pace of repair declines with age and, while exposure to a young circulation can rejuvenate fracture repair, the cell types and factors responsible for rejuvenation are unknown. Here we report that young macrophage cells produce factors that promote osteoblast differentiation of old bone marrow stromal cells. Heterochronic parabiosis exploiting young mice in which macrophages can be depleted and fractionated bone marrow transplantation experiments show that young macrophages rejuvenate fracture repair, and old macrophage cells slow healing in young mice. Proteomic analysis of the secretomes identify differential proteins secreted between old and young macrophages, such as low-density lipoprotein receptor-related protein 1 (Lrp1). Lrp1 is produced by young cells, and depleting Lrp1 abrogates the ability to rejuvenate fracture repair, while treating old mice with recombinant Lrp1 improves fracture healing. Macrophages and proteins they secrete orchestrate the fracture repair process, and young cells produce proteins that rejuvenate fracture repair in mice.


Asunto(s)
Curación de Fractura , Fracturas Óseas/fisiopatología , Macrófagos/metabolismo , Receptores de LDL/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Trasplante de Médula Ósea , Femenino , Fracturas Óseas/genética , Fracturas Óseas/metabolismo , Fracturas Óseas/terapia , Humanos , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad , Masculino , Ratones , Ratones Endogámicos C57BL , Osteoblastos/citología , Osteoblastos/metabolismo , Osteogénesis , Receptores de LDL/genética , Rejuvenecimiento , Células del Estroma/citología , Células del Estroma/metabolismo , Células del Estroma/trasplante , Proteínas Supresoras de Tumor/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA