Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 102
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Artículo en Inglés | MEDLINE | ID: mdl-38466528

RESUMEN

We identified a progenitor cell population highly enriched in samples from invasive and chemo-resistant carcinomas, characterized by a well-defined multigene signature including APOD, DCN, and LUM. This cell population has previously been labeled as consisting of inflammatory cancer-associated fibroblasts (iCAFs). The same signature characterizes naturally occurring fibro-adipogenic progenitors (FAPs) as well as stromal cells abundant in normal adipose tissue. Our analysis of human gene expression databases provides evidence that adipose stromal cells (ASCs) are recruited by tumors and undergo differentiation into CAFs during cancer progression to invasive and chemotherapy-resistant stages.

2.
Development ; 145(1)2018 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-29158445

RESUMEN

The relative abundance of thermogenic beige adipocytes and lipid-storing white adipocytes in adipose tissue underlie its metabolic activity. The roles of adipocyte progenitor cells, which express PDGFRα or PDGFRß, in adipose tissue function have remained unclear. Here, by defining the developmental timing of PDGFRα and PDGFRß expression in mouse subcutaneous and visceral adipose depots, we uncover depot specificity of pre-adipocyte delineation. We demonstrate that PDGFRα expression precedes PDGFRß expression in all subcutaneous but in only a fraction of visceral adipose stromal cells. We show that high-fat diet feeding or thermoneutrality in early postnatal development can induce PDGFRß+ lineage recruitment to generate white adipocytes. In contrast, the contribution of PDGFRß+ lineage to beige adipocytes is minimal. We provide evidence that human adipose tissue also contains distinct progenitor populations differentiating into beige or white adipocytes, depending on PDGFRß expression. Based on PDGFRα or PDGFRß deletion and ectopic expression experiments, we conclude that the PDGFRα/PDGFRß signaling balance determines progenitor commitment to beige (PDGFRα) or white (PDGFRß) adipogenesis. Our study suggests that adipocyte lineage specification and metabolism can be modulated through PDGFR signaling.


Asunto(s)
Adipocitos Beige/metabolismo , Adipocitos Blancos/metabolismo , Adipogénesis/fisiología , Diferenciación Celular/fisiología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal/fisiología , Células Madre/metabolismo , Adipocitos Beige/citología , Adipocitos Blancos/citología , Tejido Adiposo/citología , Tejido Adiposo/metabolismo , Animales , Humanos , Ratones , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Células Madre/citología
3.
Breast Cancer Res Treat ; 186(2): 273-283, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33475878

RESUMEN

PURPOSE: The purpose of this review is to clarify the association of body composition with breast cancer risk and treatment, including physiological mechanisms, and to elucidate strategies for overcoming unfavorable body composition changes that relate to breast cancer progression. METHODS: We have summarized updated knowledge regarding the mechanism of the negative association of altered body composition with breast cancer risk and treatment. We also review strategies for reversing unfavorable body composition based on the latest clinical trial results. RESULTS: Body composition changes in patients with breast cancer typically occur during menopause or as a result of chemotherapy or endocrine therapy. Dysfunction of visceral adipose tissue (VAT) in the setting of obesity underlies insulin resistance and chronic inflammation, which can lead to breast cancer development and progression. Insulin resistance and chronic inflammation are also observed in patients with breast cancer who have sarcopenia or sarcopenic obesity. Nutritional support and a personalized exercise program are the fundamental interventions for reversing unfavorable body composition. Other interventions that have been explored in specific situations include metformin, testosterone, emerging agents that directly target the adipocyte microenvironment, and bariatric surgery. CONCLUSIONS: A better understanding of the biology of body composition phenotypes is key to determining the best intervention program for patients with breast cancer.


Asunto(s)
Neoplasias de la Mama , Sarcopenia , Composición Corporal , Mama , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/epidemiología , Neoplasias de la Mama/etiología , Femenino , Humanos , Músculo Esquelético/patología , Obesidad/complicaciones , Obesidad/epidemiología , Obesidad/patología , Sarcopenia/patología , Microambiente Tumoral
4.
Stem Cells ; 37(12): 1615-1628, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31574188

RESUMEN

Although the lack of dystrophin expression in muscle myofibers is the central cause of Duchenne muscular dystrophy (DMD), accumulating evidence suggests that DMD may also be a stem cell disease. Recent studies have revealed dystrophin expression in satellite cells and demonstrated that dystrophin deficiency is directly related to abnormalities in satellite cell polarity, asymmetric division, and epigenetic regulation, thus contributing to the manifestation of the DMD phenotype. Although metabolic and mitochondrial dysfunctions have also been associated with the DMD pathophysiology profile, interestingly, the role of dystrophin with respect to stem cells dysfunction has not been elucidated. In the past few years, editing of the gene that encodes dystrophin has emerged as a promising therapeutic approach for DMD, although the effects of dystrophin restoration in stem cells have not been addressed. Herein, we describe our use of a clustered regularly interspaced short palindromic repeats/Cas9-based system to correct the dystrophin mutation in dystrophic (mdx) muscle progenitor cells (MPCs) and show that the expression of dystrophin significantly improved cellular properties of the mdx MPCs in vitro. Our findings reveal that dystrophin-restored mdx MPCs demonstrated improvements in cell proliferation, differentiation, bioenergetics, and resistance to oxidative and endoplasmic reticulum stress. Furthermore, our in vivo studies demonstrated improved transplantation efficiency of the corrected MPCs in the muscles of mdx mice. Our results indicate that changes in cellular energetics and stress resistance via dystrophin restoration enhance muscle progenitor cell function, further validating that dystrophin plays a role in stem cell function and demonstrating the potential for new therapeutic approaches for DMD. Stem Cells 2019;37:1615-1628.


Asunto(s)
Distrofina/genética , Terapia Genética/métodos , Fibras Musculares Esqueléticas/patología , Distrofia Muscular de Duchenne/terapia , Células Satélite del Músculo Esquelético/patología , Animales , Sistemas CRISPR-Cas/genética , Diferenciación Celular/genética , Polaridad Celular/fisiología , Proliferación Celular/genética , Modelos Animales de Enfermedad , Distrofina/metabolismo , Estrés del Retículo Endoplásmico/genética , Metabolismo Energético/genética , Epigénesis Genética , Edición Génica , Ratones , Ratones Endogámicos mdx , Fibras Musculares Esqueléticas/metabolismo , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patología , Estrés Oxidativo/genética , Células Madre/fisiología
5.
Cancer Metastasis Rev ; 41(3): 461-462, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35896865
6.
Stem Cells ; 35(5): 1303-1315, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28299842

RESUMEN

Electroacupuncture (EA) performed in rats and humans using limb acupuncture sites, LI-4 and LI-11, and GV-14 and GV-20 (humans) and Bai-hui (rats) increased functional connectivity between the anterior hypothalamus and the amygdala and mobilized mesenchymal stem cells (MSCs) into the systemic circulation. In human subjects, the source of the MSC was found to be primarily adipose tissue, whereas in rodents the tissue sources were considered more heterogeneous. Pharmacological disinhibition of rat hypothalamus enhanced sympathetic nervous system (SNS) activation and similarly resulted in a release of MSC into the circulation. EA-mediated SNS activation was further supported by browning of white adipose tissue in rats. EA treatment of rats undergoing partial rupture of the Achilles tendon resulted in reduced mechanical hyperalgesia, increased serum interleukin-10 levels and tendon remodeling, effects blocked in propranolol-treated rodents. To distinguish the afferent role of the peripheral nervous system, phosphoinositide-interacting regulator of transient receptor potential channels (Pirt)-GCaMP3 (genetically encoded calcium sensor) mice were treated with EA acupuncture points, ST-36 and LIV-3, and GV-14 and Bai-hui and resulted in a rapid activation of primary sensory neurons. EA activated sensory ganglia and SNS centers to mediate the release of MSC that can enhance tissue repair, increase anti-inflammatory cytokine production and provide pronounced analgesic relief. Stem Cells 2017;35:1303-1315.


Asunto(s)
Sistema Nervioso Central/citología , Electroacupuntura , Células Madre Mesenquimatosas/citología , Tendón Calcáneo/patología , Puntos de Acupuntura , Adipocitos/citología , Tejido Adiposo Pardo/citología , Tejido Adiposo Blanco/citología , Animales , Antígenos CD/metabolismo , Miembro Anterior/fisiología , Miembro Posterior/fisiología , Humanos , Hiperalgesia/terapia , Hipotálamo/citología , Interleucina-10/sangre , Macrófagos/citología , Ratones , Red Nerviosa/fisiología , Ratas , Rotura , Células Receptoras Sensoriales/metabolismo , Proteína Desacopladora 1/metabolismo
7.
Proc Natl Acad Sci U S A ; 112(27): 8403-8, 2015 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-26080435

RESUMEN

Prostate cancer antigen 3 (PCA3) is the most specific prostate cancer biomarker but its function remains unknown. Here we identify PRUNE2, a target protein-coding gene variant, which harbors the PCA3 locus, thereby classifying PCA3 as an antisense intronic long noncoding (lnc)RNA. We show that PCA3 controls PRUNE2 levels via a unique regulatory mechanism involving formation of a PRUNE2/PCA3 double-stranded RNA that undergoes adenosine deaminase acting on RNA (ADAR)-dependent adenosine-to-inosine RNA editing. PRUNE2 expression or silencing in prostate cancer cells decreased and increased cell proliferation, respectively. Moreover, PRUNE2 and PCA3 elicited opposite effects on tumor growth in immunodeficient tumor-bearing mice. Coregulation and RNA editing of PRUNE2 and PCA3 were confirmed in human prostate cancer specimens, supporting the medical relevance of our findings. These results establish PCA3 as a dominant-negative oncogene and PRUNE2 as an unrecognized tumor suppressor gene in human prostate cancer, and their regulatory axis represents a unique molecular target for diagnostic and therapeutic intervention.


Asunto(s)
Antígenos de Neoplasias/genética , Intrones/genética , Proteínas de Neoplasias/genética , Neoplasias de la Próstata/genética , ARN Largo no Codificante/genética , Proteínas Supresoras de Tumor/genética , Adenosina Desaminasa/genética , Adenosina Desaminasa/metabolismo , Animales , Antígenos de Neoplasias/metabolismo , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Células HeLa , Humanos , Immunoblotting , Células MCF-7 , Masculino , Ratones SCID , Datos de Secuencia Molecular , Proteínas de Neoplasias/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Unión Proteica , Interferencia de ARN , Precursores del ARN/genética , Precursores del ARN/metabolismo , ARN Largo no Codificante/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Supresoras de Tumor/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
8.
Stem Cells ; 34(1): 174-90, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26381424

RESUMEN

Adipose stromal cells (ASC) are mesenchymal adipocyte progenitors that reside in the peri-endothelium of fat tissue. ASC mobilization and migration accompany white adipose tissue (WAT) remodeling and pathological conditions. Mechanisms regulating ASC trafficking are largely unknown. We previously reported that binding of the matricellular protein secreted protein acidic and rich in cysteine (SPARC) to ß1 integrin on ASC surface induces their motility. Here, we show that SPARC is required for ASC mobilization. We report two SPARC proteolytic isoforms, C-SPARC (lacking the N terminus) and N-SPARC (lacking the C terminus), generated in mesenteric WAT of obese mice. C-SPARC, but not N-SPARC, binds to ß1 integrin on ASC, while N-SPARC preferentially binds to the extracellular matrix (ECM) and blocks ECM/integrin interaction. Interestingly, both C-SPARC and N-SPARC induce ASC deadhesion from the ECM, which is associated with modulation of integrin-dependent FAK-ERK signaling and integrin-independent ILK-Akt signaling. We show that these SPARC isoforms, acting on ASC through distinct mechanisms, have an additive effect in inducing ASC migration.


Asunto(s)
Tejido Adiposo/patología , Obesidad/metabolismo , Obesidad/patología , Osteonectina/metabolismo , Proteolisis , Células 3T3-L1 , Secuencia de Aminoácidos , Animales , Adhesión Celular , Movimiento Celular , Matriz Extracelular/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Células HEK293 , Humanos , Integrina alfa5 , Integrina beta1/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Datos de Secuencia Molecular , Osteonectina/química , Unión Proteica , Isoformas de Proteínas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Células del Estroma/metabolismo
9.
Mol Ther ; 24(1): 34-40, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26316391

RESUMEN

Progression of many cancers is associated with tumor infiltration by mesenchymal stromal cells (MSC). Adipose stromal cells (ASC) are MSC that serve as adipocyte progenitors and endothelium-supporting cells in white adipose tissue (WAT). Clinical and animal model studies indicate that ASC mobilized from WAT are recruited by tumors. Direct evidence for ASC function in tumor microenvironment has been lacking due to unavailability of approaches to specifically inactivate these cells. Here, we investigate the effects of a proteolysis-resistant targeted hunter-killer peptide D-WAT composed of a cyclic domain CSWKYWFGEC homing to ASC and of a proapoptotic domain KLAKLAK2. Using mouse bone marrow transplantation models, we show that D-WAT treatment specifically depletes tumor stromal and perivascular cells without directly killing malignant cells or tumor-infiltrating leukocytes. In several mouse carcinoma models, targeted ASC cytoablation reduced tumor vascularity and cell proliferation resulting in hemorrhaging, necrosis, and suppressed tumor growth. We also validated a D-WAT derivative with a proapoptotic domain KFAKFAK2 that was found to have an improved cytoablative activity. Our results for the first time demonstrate that ASC, recruited as a component of tumor microenvironment, support cancer progression. We propose that drugs targeting ASC can be developed as a combination therapy complementing conventional cancer treatments.


Asunto(s)
Tejido Adiposo/citología , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Melanoma Experimental/tratamiento farmacológico , Células Madre Mesenquimatosas/efectos de los fármacos , Neovascularización Patológica/tratamiento farmacológico , Péptidos/administración & dosificación , Animales , Trasplante de Médula Ósea , Carcinoma Pulmonar de Lewis/patología , Diferenciación Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Melanoma Experimental/patología , Células Madre Mesenquimatosas/citología , Ratones , Péptidos/farmacología , Microambiente Tumoral/efectos de los fármacos
10.
Am J Physiol Endocrinol Metab ; 311(6): E952-E963, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27729337

RESUMEN

Endotrophin is a cleavage product of collagen 6 (Col6) in adipose tissue (AT). Previously, we demonstrated that endotrophin serves as a costimulator to trigger fibrosis and inflammation within the unhealthy AT milieu. However, how endotrophin affects lipid storage and breakdown in AT and how different cell types in AT respond to endotrophin stimulation remain unknown. In the current study, by using a doxycycline-inducible mouse model, we observed significant upregulation of adipogenic genes in the white AT (WAT) of endotrophin transgenic mice. We further showed that the mice exhibited inhibited lipolysis and accelerated hypertrophy and hyperplasia in WAT. To investigate the effects of endotrophin in vitro, we incubated different cell types from AT with conditioned medium from endotrophin-overexpressing 293T cells. We found that endotrophin activated multiple pathological pathways in different cell types. Particularly in 3T3-L1 adipocytes, endotrophin triggered a fibrotic program by upregulating collagen genes and promoted abnormal lipid accumulation by downregulating hormone-sensitive lipolysis gene and decreasing HSL phosphorylation levels. In macrophages isolated from WAT, endotrophin stimulated higher expression of the collagen-linking enzyme lysyl oxidase and M1 proinflammatory marker genes. In the stromal vascular fraction isolated from WAT, endotrophin induced upregulation of both profibrotic and proinflammatory genes. In conclusion, our study provides a new perspective on the effect of endotrophin in abnormal lipid accumulation and a mechanistic insight into the roles played by adipocytes and a variety of other cell types in AT in shaping the unhealthy microenvironment upon endotrophin treatment.


Asunto(s)
Adipogénesis/genética , Tejido Adiposo Blanco/metabolismo , Tejido Adiposo/citología , Colágeno Tipo VI/genética , Fibrosis/genética , Inflamación/genética , Lipólisis/genética , Fragmentos de Péptidos/genética , Células 3T3-L1 , Tejido Adiposo Blanco/patología , Animales , Colágeno/genética , Células HEK293 , Humanos , Hiperplasia , Hipertrofia , Macrófagos/inmunología , Ratones , Ratones Transgénicos , Fosforilación/genética , Proteína-Lisina 6-Oxidasa/genética , Esterol Esterasa/genética , Regulación hacia Arriba
11.
Proc Natl Acad Sci U S A ; 109(5): 1637-42, 2012 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-22307623

RESUMEN

Processes that promote cancer progression such as angiogenesis require a functional interplay between malignant and nonmalignant cells in the tumor microenvironment. The metalloprotease aminopeptidase N (APN; CD13) is often overexpressed in tumor cells and has been implicated in angiogenesis and cancer progression. Our previous studies of APN-null mice revealed impaired neoangiogenesis in model systems without cancer cells and suggested the hypothesis that APN expressed by nonmalignant cells might promote tumor growth. We tested this hypothesis by comparing the effects of APN deficiency in allografted malignant (tumor) and nonmalignant (host) cells on tumor growth and metastasis in APN-null mice. In two independent tumor graft models, APN activity in both the tumors and the host cells cooperate to promote tumor vascularization and growth. Loss of APN expression by the host and/or the malignant cells also impaired lung metastasis in experimental mouse models. Thus, cooperation in APN expression by both cancer cells and nonmalignant stromal cells within the tumor microenvironment promotes angiogenesis, tumor growth, and metastasis.


Asunto(s)
Antígenos CD13/metabolismo , Neoplasias Pulmonares/enzimología , Animales , Antígenos CD13/genética , Línea Celular Tumoral , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
12.
Curr Opin Oncol ; 26(6): 545-50, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25279961

RESUMEN

PURPOSE OF REVIEW: There is emerging evidence that obesity is associated with an increase in the incidence, severity, and mortality from different types of cancer, including postmenopausal breast cancer. Here, we discuss the role of white adipose tissue (WAT) cells and of related soluble factors in the local and metastatic growth of this neoplastic disease. Moreover, we discuss the recent increase in the use of WAT-derived progenitor cells in breast cancer patients to enhance the quality of breast reconstruction and the related risks. RECENT FINDINGS: In several murine models, WAT cells and progenitors were found to have cooperative roles in promoting local breast cancer. Moreover, they were found to contribute to adipocytes and pericytes supporting the cancer vasculature, and stimulated the metastatic progression of breast cancer. There are some clinically retrospective data showing a significant increase in the frequency of intraepithelial neoplasia in patients who received a lipofilling procedure for breast reconstruction compared with controls. SUMMARY: Preclinical models and clinical studies are urgently needed to investigate how to inhibit the tumor-promoting activity of WAT cells and progenitors. The risks associated with the use of WAT cells for breast reconstructions should be better investigated retrospectively and prospectively.


Asunto(s)
Tejido Adiposo Blanco , Neoplasias de la Mama/etiología , Células Madre , Animales , Neoplasias de la Mama/patología , Femenino , Humanos , Inflamación/metabolismo , Obesidad/complicaciones , Factores de Riesgo
13.
Proc Natl Acad Sci U S A ; 108(46): 18637-42, 2011 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-22049339

RESUMEN

Molecules differentially expressed in blood vessels among organs or between damaged and normal tissues, are attractive therapy targets; however, their identification within the human vasculature is challenging. Here we screened a peptide library in cancer patients to uncover ligand-receptors common or specific to certain vascular beds. Surveying ~2.35 x 10(6) motifs recovered from biopsies yielded a nonrandom distribution, indicating that systemic tissue targeting is feasible. High-throughput analysis by similarity search, protein arrays, and affinity chromatography revealed four native ligand-receptors, three of which were previously unrecognized. Two are shared among multiple tissues (integrin α4/annexin A4 and cathepsin B/apolipoprotein E3) and the other two have a restricted and specific distribution in normal tissue (prohibitin/annexin A2 in white adipose tissue) or cancer (RAGE/leukocyte proteinase-3 in bone metastases). These findings provide vascular molecular markers for biotechnology and medical applications.


Asunto(s)
Vasos Sanguíneos/metabolismo , Médula Ósea/metabolismo , Neoplasias/metabolismo , Secuencias de Aminoácidos , Anexina A4/biosíntesis , Apolipoproteína E3/biosíntesis , Biopsia , Catepsina B/biosíntesis , Regulación Neoplásica de la Expresión Génica , Humanos , Integrina alfa4/biosíntesis , Ligandos , Neovascularización Patológica , Obesidad/metabolismo , Biblioteca de Péptidos
14.
Cancer Res ; 84(5): 648-649, 2024 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-38437636

RESUMEN

Cancer aggressiveness has been linked with obesity, and studies have shown that adipose tissue can enhance cancer progression. In this issue of Cancer Research, Hosni and colleagues discover a paracrine mechanism mediated by adipocyte precursor cells through which urothelial carcinomas become resistant to erdafitinib, a recently approved therapy inhibiting fibroblast growth factor receptors (FGFR). They identified neuregulin 1 (NRG1) secreted by adipocyte precursor cells as an activator of HER3 signaling that enables resistance. The NRG1-mediated FGFR inhibitor resistance was amenable to intervention with pertuzumab, an antibody blocking the NRG1/HER3 axis. To investigate the nature of the resistance-associated NRG1-expressing cells in human patients, the authors analyzed published single-cell RNA sequencing data and observed that such cells appear in a cluster assigned as inflammatory cancer-associated fibroblasts (iCAF). Notably, the gene signature corresponding to these CAFs is highly similar to that shared by adipose stromal cells (ASC) in fat tissue and fibro-adipogenic progenitors (FAP) in skeletal muscle of cancer-free individuals. Because fibroblasts with the ASC/FAP signature are enriched in various carcinomas, it is possible that the paracrine signaling conferred by NRG1 is a pan-cancer mechanism of FGFR inhibitor resistance and tumor aggressiveness. See related article by Hosni et al., p. 725.


Asunto(s)
Fibroblastos Asociados al Cáncer , Carcinoma de Células Transicionales , Humanos , Adipocitos , Tejido Adiposo , Células del Estroma
15.
Aging Cell ; 23(6): e14138, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38475941

RESUMEN

It has remained unclear how aging of endothelial cells (EC) contributes to pathophysiology of individual organs. Cell senescence results in part from inactivation of telomerase (TERT). Here, we analyzed mice with Tert knockout specifically in EC. Tert loss in EC induced transcriptional changes indicative of senescence and tissue hypoxia in EC and in other cells. We demonstrate that EC-Tert-KO mice have leaky blood vessels. The blood-brain barrier of EC-Tert-KO mice is compromised, and their cognitive function is impaired. EC-Tert-KO mice display reduced muscle endurance and decreased expression of enzymes responsible for oxidative metabolism. Our data indicate that Tert-KO EC have reduced mitochondrial content and function, which results in increased dependence on glycolysis. Consistent with this, EC-Tert-KO mice have metabolism changes indicative of increased glucose utilization. In EC-Tert-KO mice, expedited telomere attrition is observed for EC of adipose tissue (AT), while brain and skeletal muscle EC have normal telomere length but still display features of senescence. Our data indicate that the loss of Tert causes EC senescence in part through a telomere length-independent mechanism undermining mitochondrial function. We conclude that EC-Tert-KO mice is a model of expedited vascular senescence recapitulating the hallmarks aging, which can be useful for developing revitalization therapies.


Asunto(s)
Envejecimiento , Senescencia Celular , Células Endoteliales , Ratones Noqueados , Telomerasa , Telómero , Animales , Telomerasa/metabolismo , Telomerasa/genética , Senescencia Celular/genética , Envejecimiento/metabolismo , Ratones , Células Endoteliales/metabolismo , Telómero/metabolismo , Telómero/genética , Mitocondrias/metabolismo
16.
Mol Metab ; 84: 101933, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38583571

RESUMEN

OBJECTIVE: Alström Syndrome (AS), caused by biallelic ALMS1 mutations, includes obesity with disproportionately severe insulin resistant diabetes, dyslipidemia, and fatty liver. Prior studies suggest that hyperphagia is accounted for by loss of ALMS1 function in hypothalamic neurones, whereas disproportionate metabolic complications may be due to impaired adipose tissue expandability. We tested this by comparing the metabolic effects of global and mesenchymal stem cell (MSC)-specific Alms1 knockout. METHODS: Global Alms1 knockout (KO) mice were generated by crossing floxed Alms1 and CAG-Cre mice. A Pdgfrα-Cre driver was used to abrogate Alms1 function selectively in MSCs and their descendants, including preadipocytes. We combined metabolic phenotyping of global and Pdgfrα+ Alms1-KO mice on a 45% fat diet with measurements of body composition and food intake, and histological analysis of metabolic tissues. RESULTS: Assessed on 45% fat diet to promote adipose expansion, global Alms1 KO caused hyperphagia, obesity, insulin resistance, dyslipidaemia, and fatty liver. Pdgfrα-cre driven KO of Alms1 (MSC KO) recapitulated insulin resistance, fatty liver, and dyslipidaemia in both sexes. Other phenotypes were sexually dimorphic: increased fat mass was only present in female Alms1 MSC KO mice. Hyperphagia was not evident in male Alms1 MSC KO mice, but was found in MSC KO females, despite no neuronal Pdgfrα expression. CONCLUSIONS: Mesenchymal deletion of Alms1 recapitulates metabolic features of AS, including fatty liver. This confirms a key role for Alms1 in the adipose lineage, where its loss is sufficient to cause systemic metabolic effects and damage to remote organs. Hyperphagia in females may depend on Alms1 deficiency in oligodendrocyte precursor cells rather than neurones. AS should be regarded as a forme fruste of lipodystrophy.


Asunto(s)
Síndrome de Alstrom , Células Madre Mesenquimatosas , Ratones Noqueados , Animales , Ratones , Masculino , Femenino , Células Madre Mesenquimatosas/metabolismo , Síndrome de Alstrom/metabolismo , Síndrome de Alstrom/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Resistencia a la Insulina , Hígado Graso/metabolismo , Hígado Graso/genética , Obesidad/metabolismo , Obesidad/genética , Hiperfagia/metabolismo , Hiperfagia/genética , Tejido Adiposo/metabolismo , Ratones Endogámicos C57BL , Composición Corporal
17.
Diabetes ; 73(5): 701-712, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38320268

RESUMEN

Bile acids (BAs) are pleiotropic regulators of metabolism. Elevated levels of hepatic and circulating BAs improve energy metabolism in peripheral organs, but the precise mechanisms underlying the metabolic benefits and harm still need to be fully understood. In the current study, we identified orosomucoid 2 (ORM2) as a liver-secreted hormone (i.e., hepatokine) induced by BAs and investigated its role in BA-induced metabolic improvements in mouse models of diet-induced obesity. Contrary to our expectation, under a high-fat diet (HFD), our Orm2 knockout (Orm2-KO) exhibited a lean phenotype compared with C57BL/6J control, partly due to the increased energy expenditure. However, when challenged with a HFD supplemented with cholic acid, Orm2-KO eliminated the antiobesity effect of BAs, indicating that ORM2 governs BA-induced metabolic improvements. Moreover, hepatic ORM2 overexpression partially replicated BA effects by enhancing insulin sensitivity. Mechanistically, ORM2 suppressed interferon-γ/STAT1 activities in inguinal white adipose tissue depots, forming the basis for anti-inflammatory effects of BAs and improving glucose homeostasis. In conclusion, our study provides new insights into the molecular mechanisms of BA-induced liver-adipose cross talk through ORM2 induction.


Asunto(s)
Ácidos y Sales Biliares , Orosomucoide , Ratones , Animales , Ácidos y Sales Biliares/metabolismo , Orosomucoide/metabolismo , Orosomucoide/farmacología , Ratones Endogámicos C57BL , Obesidad/genética , Obesidad/metabolismo , Hígado/metabolismo , Dieta Alta en Grasa/efectos adversos
18.
Biochim Biophys Acta ; 1826(1): 209-14, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22546620

RESUMEN

Despite recent evidence of the cancer-promoting role of adipose tissue-derived progenitor and differentiated cells, the use of lipotransfer for tissue/organ reconstruction after surgical removal of cancer is increasing worldwide. Here we discuss in a multidisciplinary fashion the preclinical data connecting obesity, adipose cells and cancer progression, as well as the clinical data concerning safety of lipotransfer procedures in cancer patients. A roadmap towards a more rationale use of lipotransfer in oncology is urgently needed and should include preclinical studies to dissect the roles of different adipose tissue-derived cells, the evaluation of drugs currently candidate to inhibit the interaction between adipose and tumor cells, and carefully designed clinical trials to investigate the safety of lipotransfer procedures in cancer patients.


Asunto(s)
Adipocitos/patología , Tejido Adiposo/patología , Tejido Adiposo/trasplante , Neoplasias/etiología , Procedimientos de Cirugía Plástica/efectos adversos , Adipocitos/citología , Tejido Adiposo/citología , Humanos , Neoplasias/cirugía , Procedimientos de Cirugía Plástica/métodos
19.
Small ; 9(9-10): 1696-702, 2013 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-23166049

RESUMEN

In vitro toxicity assays based on the evaluation and retention of advanced and specific cellular functions are proposed to investigate mesoporous silicon nanovectors. This approach provides greater insight compared to simple cellular viability and toxicity assays. Electron microscopy images demonstrate internalized nanovectors altering the curvature of the nuclear envelope with minimal effect on viability or biological function.


Asunto(s)
Supervivencia Celular , Endocitosis , Nanopartículas , Células Cultivadas , Humanos
20.
Biometrics ; 69(1): 174-83, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23339534

RESUMEN

We discuss inference for a human phage display experiment with three stages. The data are tripeptide counts by tissue and stage. The primary aim of the experiment is to identify ligands that bind with high affinity to a given tissue. We formalize the research question as inference about the monotonicity of mean counts over stages. The inference goal is then to identify a list of peptide-tissue pairs with significant increase over stages. We use a semiparametric Dirichlet process mixture of Poisson model. The posterior distribution under this model allows the desired inference about the monotonicity of mean counts. However, the desired inference summary as a list of peptide-tissue pairs with significant increase involves a massive multiplicity problem. We consider two alternative approaches to address this multiplicity issue. First we propose an approach based on the control of the posterior expected false discovery rate. We notice that the implied solution ignores the relative size of the increase. This motivates a second approach based on a utility function that includes explicit weights for the size of the increase.


Asunto(s)
Teorema de Bayes , Modelos Estadísticos , Biblioteca de Péptidos , Tejido Adiposo/metabolismo , Médula Ósea/metabolismo , Simulación por Computador , Humanos , Masculino , Cadenas de Markov , Método de Montecarlo , Oligopéptidos/metabolismo , Próstata/metabolismo , Piel/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA