Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Gastroenterology ; 166(3): 437-449, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37995867

ABSTRACT

BACKGROUND & AIMS: RET tyrosine kinase is necessary for enteric nervous system development. Loss-of-function RET mutations cause Hirschsprung disease (HSCR), in which infants are born with aganglionic bowel. Despite surgical correction, patients with HSCR often experience chronic defecatory dysfunction and enterocolitis, suggesting that RET is important after development. To test this hypothesis, we determined the location of postnatal RET and its significance in gastrointestinal (GI) motility. METHODS: RetCFP/+ mice and human transcriptional profiling data were studied to identify the enteric neuronal and epithelial cells that express RET. To determine whether RET regulates gut motility in vivo, genetic, and pharmacologic approaches were used to disrupt RET in all RET-expressing cells, a subset of enteric neurons, or intestinal epithelial cells. RESULTS: Distinct subsets of enteric neurons and enteroendocrine cells expressed RET in the adult intestine. RET disruption in the epithelium, rather than in enteric neurons, slowed GI motility selectively in male mice. RET kinase inhibition phenocopied this effect. Most RET+ epithelial cells were either enterochromaffin cells that release serotonin or L-cells that release peptide YY (PYY) and glucagon-like peptide 1 (GLP-1), both of which can alter motility. RET kinase inhibition exaggerated PYY and GLP-1 release in a nutrient-dependent manner without altering serotonin secretion in mice and human organoids. PYY receptor blockade rescued dysmotility in mice lacking epithelial RET. CONCLUSIONS: RET signaling normally limits nutrient-dependent peptide release from L-cells and this activity is necessary for normal intestinal motility in male mice. These effects could contribute to dysmotility in HSCR, which predominantly affects males, and uncovers a mechanism that could be targeted to treat post-prandial GI dysfunction.


Subject(s)
Enteric Nervous System , Hirschsprung Disease , Infant , Humans , Male , Mice , Animals , Peptide YY , Serotonin , Hirschsprung Disease/genetics , Enteroendocrine Cells , Intestine, Small , Glucagon-Like Peptide 1 , Proto-Oncogene Proteins c-ret/genetics
2.
Dev Cell ; 59(2): 244-261.e6, 2024 Jan 22.
Article in English | MEDLINE | ID: mdl-38154460

ABSTRACT

WNT morphogens trigger signaling pathways fundamental for embryogenesis, regeneration, and cancer. WNTs are modified with palmitoleate, which is critical for binding Frizzled (FZD) receptors and activating signaling. However, it is unknown how WNTs are released and spread from cells, given their strong lipid-dependent membrane attachment. We demonstrate that secreted FZD-related proteins and WNT inhibitory factor 1 are WNT carriers, potently releasing lipidated WNTs and forming active soluble complexes. WNT release occurs by direct handoff from the membrane protein WNTLESS to the carriers. In turn, carriers donate WNTs to glypicans and FZDs involved in WNT reception and to the NOTUM hydrolase, which antagonizes WNTs by lipid moiety removal. WNT transfer from carriers to FZDs is greatly facilitated by glypicans that serve as essential co-receptors in Wnt signaling. Thus, an extracellular network of carriers dynamically controls secretion, posttranslational regulation, and delivery of WNT morphogens, with important practical implications for regenerative medicine.


Subject(s)
Glypicans , Wnt Proteins , Wnt Proteins/metabolism , Glypicans/metabolism , Wnt Signaling Pathway , Embryonic Development , Lipids , Frizzled Receptors/chemistry , Frizzled Receptors/metabolism
3.
Gut ; 72(5): 870-881, 2023 05.
Article in English | MEDLINE | ID: mdl-35537812

ABSTRACT

OBJECTIVE: Intestinal barrier loss is a Crohn's disease (CD) risk factor. This may be related to increased expression and enzymatic activation of myosin light chain kinase 1 (MLCK1), which increases intestinal paracellular permeability and correlates with CD severity. Moreover, preclinical studies have shown that MLCK1 recruitment to cell junctions is required for tumour necrosis factor (TNF)-induced barrier loss as well as experimental inflammatory bowel disease progression. We sought to define mechanisms of MLCK1 recruitment and to target this process pharmacologically. DESIGN: Protein interactions between FK506 binding protein 8 (FKBP8) and MLCK1 were assessed in vitro. Transgenic and knockout intestinal epithelial cell lines, human intestinal organoids, and mice were used as preclinical models. Discoveries were validated in biopsies from patients with CD and control subjects. RESULTS: MLCK1 interacted specifically with the tacrolimus-binding FKBP8 PPI domain. Knockout or dominant negative FKBP8 expression prevented TNF-induced MLCK1 recruitment and barrier loss in vitro. MLCK1-FKBP8 binding was blocked by tacrolimus, which reversed TNF-induced MLCK1-FKBP8 interactions, MLCK1 recruitment and barrier loss in vitro and in vivo. Biopsies of patient with CD demonstrated increased numbers of MLCK1-FKBP8 interactions at intercellular junctions relative to control subjects. CONCLUSION: Binding to FKBP8, which can be blocked by tacrolimus, is required for MLCK1 recruitment to intercellular junctions and downstream events leading to immune-mediated barrier loss. The observed increases in MLCK1 activity, MLCK1 localisation at cell junctions and perijunctional MLCK1-FKBP8 interactions in CD suggest that targeting this process may be therapeutic in human disease. These new insights into mechanisms of disease-associated barrier loss provide a critical foundation for therapeutic exploitation of FKBP8-MLCK1 interactions.


Subject(s)
Crohn Disease , Animals , Humans , Mice , Caco-2 Cells , Crohn Disease/drug therapy , Crohn Disease/metabolism , Intestinal Mucosa/metabolism , Mice, Knockout , Myosin-Light-Chain Kinase/metabolism , Tacrolimus/pharmacology , Tacrolimus Binding Proteins/metabolism , Tight Junctions/physiology , Tumor Necrosis Factor-alpha/metabolism
4.
Nat Commun ; 13(1): 6786, 2022 11 09.
Article in English | MEDLINE | ID: mdl-36351897

ABSTRACT

Toxin B (TcdB) is a major exotoxin responsible for diseases associated with Clostridioides difficile infection. Its sequence variations among clinical isolates may contribute to the difficulty in developing effective therapeutics. Here, we investigate receptor-binding specificity of major TcdB subtypes (TcdB1 to TcdB12). We find that representative members of subtypes 2, 4, 7, 10, 11, and 12 do not recognize the established host receptor, frizzled proteins (FZDs). Using a genome-wide CRISPR-Cas9-mediated screen, we identify tissue factor pathway inhibitor (TFPI) as a host receptor for TcdB4. TFPI is recognized by a region in TcdB4 that is homologous to the FZD-binding site in TcdB1. Analysis of 206 TcdB variant sequences reveals a set of six residues within this receptor-binding site that defines a TFPI binding-associated haplotype (designated B4/B7) that is present in all TcdB4 members, a subset of TcdB7, and one member of TcdB2. Intragenic micro-recombination (IR) events have occurred around this receptor-binding region in TcdB7 and TcdB2 members, resulting in either TFPI- or FZD-binding capabilities. Introduction of B4/B7-haplotype residues into TcdB1 enables dual recognition of TFPI and FZDs. Finally, TcdB10 also recognizes TFPI, although it does not belong to the B4/B7 haplotype, and shows species selectivity: it recognizes TFPI of chicken and to a lesser degree mouse, but not human, dog, or cattle versions. These findings identify TFPI as a TcdB receptor and reveal IR-driven changes on receptor-specificity among TcdB variants.


Subject(s)
Bacterial Toxins , Clostridioides difficile , Animals , Cattle , Dogs , Mice , Bacterial Proteins/metabolism , Bacterial Toxins/chemistry , Clostridioides difficile/genetics , Recombination, Genetic , Humans
5.
Nat Commun ; 13(1): 261, 2022 01 11.
Article in English | MEDLINE | ID: mdl-35017529

ABSTRACT

Enteroendocrine (EE) cells are the most abundant hormone-producing cells in humans and are critical regulators of energy homeostasis and gastrointestinal function. Challenges in converting human intestinal stem cells (ISCs) into functional EE cells, ex vivo, have limited progress in elucidating their role in disease pathogenesis and in harnessing their therapeutic potential. To address this, we employed small molecule targeting of the endocannabinoid receptor signaling pathway, JNK, and FOXO1, known to mediate endodermal development and/or hormone production, together with directed differentiation of human ISCs from the duodenum and rectum. We observed marked induction of EE cell differentiation and gut-derived expression and secretion of SST, 5HT, GIP, CCK, GLP-1 and PYY upon treatment with various combinations of three small molecules: rimonabant, SP600125 and AS1842856. Robust differentiation strategies capable of driving human EE cell differentiation is a critical step towards understanding these essential cells and the development of cell-based therapeutics.


Subject(s)
Cell Differentiation/drug effects , Cell Differentiation/physiology , Enteroendocrine Cells/drug effects , Enteroendocrine Cells/metabolism , Stem Cells/drug effects , Stem Cells/metabolism , Anthracenes/pharmacology , Chromogranin A/metabolism , Endocannabinoids/pharmacology , Glucagon-Like Peptide 1/metabolism , Humans , Intestinal Mucosa/metabolism , Peptide YY/metabolism , Quinolones/pharmacology , Rimonabant/pharmacology , Signal Transduction , Somatostatin/metabolism , Transcription Factors/metabolism
6.
J Clin Endocrinol Metab ; 107(4): e1610-e1619, 2022 03 24.
Article in English | MEDLINE | ID: mdl-34791361

ABSTRACT

CONTEXT: Pseudohypoparathyroidism type Ib (PHP1B) is characterized by hypocalcemia and hyperphosphatemia due to parathyroid hormone resistance in the proximal renal tubules. Maternal pathogenic STX16/GNAS variants leading to maternal epigenetic GNAS changes impair expression of the stimulatory G protein alpha-subunit (Gsα) thereby causing autosomal dominant PHP1B. In contrast, genetic defects responsible for sporadic PHP1B (sporPHP1B) remain mostly unknown. OBJECTIVE: Determine whether PHP1B encountered after in vitro fertilization (IVF) or intracytoplasmic sperm injection (ICSI) causes GNAS remethylation defects similar to those in sporPHP1B. DESIGN: Retrospective analysis. RESULTS: Nine among 36 sporPHP1B patients investigated since 2000, all with loss of methylation (LOM) at the 3 maternal GNAS differentially methylated regions (DMRs) and gain of methylation at the paternal NESP DMR, had been conceived through IVF or ICSI. Besides abnormal GNAS methylation, IVF/ICSI PHP1B cases revealed no additional imprinting defects. Three of these PHP1B patients have dizygotic twins, and 4 have IVF/ICSI-conceived siblings, all with normal GNAS methylation; 2 unaffected younger siblings were conceived naturally. CONCLUSION: Sporadic and IVF/ICSI-conceived PHP1B patients revealed indistinguishable epigenetic changes at all 4 GNAS DMRs, thus suggesting a similar underlying disease mechanism. Given that remethylation at the 3 maternal DMRs occurs during oogenesis, male factors are unlikely to cause LOM postfertilization. Instead, at least some of the sporPHP1B variants could be caused by a defect or defects in an oocyte-expressed gene that is required for fertility and for re-establishing maternal GNAS methylation imprints. It remains uncertain, however, whether the lack of GNAS remethylation alone and the resulting reduction in Gsα expression is sufficient to impair oocyte maturation.


Subject(s)
Chromogranins , Pseudohypoparathyroidism , Chromogranins/genetics , DNA Methylation , GTP-Binding Protein alpha Subunits, Gs/genetics , Humans , Male , Oogenesis , Pseudohypoparathyroidism/genetics , Retrospective Studies , Pseudohypoparathyroidism
7.
BMJ Case Rep ; 12(3)2019 Mar 20.
Article in English | MEDLINE | ID: mdl-30898952

ABSTRACT

11-year old twin boy found to have idiopathic precocious puberty after routine well-child examination revealed discordant pubertal growth between the two brothers.


Subject(s)
Puberty, Precocious/diagnosis , Twins, Dizygotic , Acne Vulgaris , Child , Growth Charts , Hair , Humans , Male , Physical Examination
9.
PLoS One ; 11(3): e0152129, 2016.
Article in English | MEDLINE | ID: mdl-27015423

ABSTRACT

Exercise decreases adiposity and improves metabolic health; however, the physiological and molecular underpinnings of these phenomena remain unknown. Here, we investigate the effect of endurance training on adipose progenitor lineage commitment. Using mice with genetically labeled adipose progenitors, we show that these cells react to exercise by decreasing their proliferation and differentiation potential. Analyses of mouse models that mimic the skeletal muscle adaptation to exercise indicate that muscle, in a non-autonomous manner, regulates adipose progenitor homeostasis, highlighting a role for muscle-derived secreted factors. These findings support a humoral link between skeletal muscle and adipose progenitors and indicate that manipulation of adipose stem cell function may help address obesity and diabetes.


Subject(s)
Adipose Tissue/cytology , Muscle, Skeletal/physiology , Physical Conditioning, Animal , Stem Cells/cytology , 3T3-L1 Cells , Adaptation, Physiological , Adipocytes/cytology , Adipose Tissue/metabolism , Animals , Cell Differentiation , Cell Line , Cell Lineage , Cell Proliferation , Culture Media, Conditioned , Diabetes Mellitus/metabolism , Glucose Tolerance Test , Green Fluorescent Proteins/metabolism , Homeostasis , Male , Mice , Mice, Transgenic , Obesity/metabolism , Oligonucleotide Array Sequence Analysis , Physical Endurance/physiology , Real-Time Polymerase Chain Reaction , Thrombospondins/metabolism
10.
Nat Commun ; 5: 5196, 2014 Oct 21.
Article in English | MEDLINE | ID: mdl-25330806

ABSTRACT

Oestrogen, often via oestrogen receptor alpha (ERα) signalling, regulates metabolic physiology, highlighted by post-menopausal temperature dysregulation (hot flashes), glucose intolerance, increased appetite and reduced metabolic rate. Here we show that ERα signalling has a role in adipose lineage specification in mice. ERα regulates adipose progenitor identity and potency, promoting white adipogenic lineage commitment. White adipose progenitors lacking ERα reprogramme and enter into smooth muscle and brown adipogenic fates. Mechanistic studies highlight a TGFß programme involved in progenitor reprogramming downstream of ERα signalling. The observed reprogramming has profound metabolic outcomes; both female and male adipose-lineage ERα-mutant mice are lean, have improved glucose sensitivity and are resistant to weight gain on a high-fat diet. Further, they are hypermetabolic, hyperphagic and hyperthermic, all consistent with a brown phenotype. Together, these findings indicate that ERα cell autonomously regulates adipose lineage commitment, brown fat and smooth muscle cell formation, and systemic metabolism, in a manner relevant to prevalent metabolic diseases.


Subject(s)
Adipose Tissue, Brown/cytology , Cell Differentiation , Estrogens/metabolism , Myocytes, Smooth Muscle/cytology , Signal Transduction , Stem Cells/metabolism , Adipose Tissue/cytology , Adipose Tissue, White/cytology , Animals , Cell Lineage , Cell Proliferation , Cell Separation , Estrogen Receptor alpha/metabolism , Female , Flow Cytometry , Green Fluorescent Proteins/metabolism , Male , Mice , Mice, Mutant Strains , Mutation , Neovascularization, Physiologic , Phenotype , Random Allocation , Stem Cells/cytology , Transforming Growth Factor beta/metabolism
11.
Mol Metab ; 2(3): 227-42, 2013.
Article in English | MEDLINE | ID: mdl-24049737

ABSTRACT

Our data demonstrate that estrogens, estrogen receptor-α (ERα), and estrogen receptor-ß (ERß) regulate adipose tissue distribution, inflammation, fibrosis, and glucose homeostasis, by determining that αERKO mice have increased adipose tissue inflammation and fibrosis prior to obesity onset. Selective deletion of adipose tissue ERα in adult mice using a novel viral vector technology recapitulated the findings in the total body ERα null mice. Generation of a novel mouse model, lacking ERα specifically from adipocytes (AdipoERα), demonstrated increased markers of fibrosis and inflammation, especially in the males. Additionally, we found that the beneficial effects of estrogens on adipose tissue require adipocyte ERα. Lastly, we determined the role of ERß in regulating inflammation and fibrosis, by breeding the AdipoERα into the ßERKO background and found that in the absence of adipocyte ERα, ERß has a protective role. These data suggest that adipose tissue and adipocyte ERα protects against adiposity, inflammation, and fibrosis in both males and females.

12.
Development ; 140(19): 3939-49, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24046315

ABSTRACT

Adipose tissue is formed at stereotypic times and locations in a diverse array of organisms. Once formed, the tissue is dynamic, responding to homeostatic and external cues and capable of a 15-fold expansion. The formation and maintenance of adipose tissue is essential to many biological processes and when perturbed leads to significant diseases. Despite this basic and clinical significance, understanding of the developmental biology of adipose tissue has languished. In this Review, we highlight recent efforts to unveil adipose developmental cues, adipose stem cell biology and the regulators of adipose tissue homeostasis and dynamism.


Subject(s)
Adipose Tissue/cytology , Adipocytes/cytology , Animals , Cell Differentiation/physiology , Humans , Stem Cell Niche/physiology , Stem Cells/cytology
13.
Cell Metab ; 15(4): 492-504, 2012 Apr 04.
Article in English | MEDLINE | ID: mdl-22482731

ABSTRACT

Adipose tissues provide circulating nutrients and hormones. We present in vivo mouse studies highlighting roles for Wnt signals in both aspects of metabolism. ß-catenin activation in PPARγ-expressing fat progenitors (PBCA) decreased fat mass and induced fibrotic replacement of subcutaneous fat specifically. In spite of lipodystrophy, PBCA mice did not develop the expected diabetes and hepatosteatosis, but rather exhibited improved glucose metabolism and normal insulin sensitivity. Glucose uptake was increased in muscle independently of insulin, associated with cell-surface translocation of glucose transporters and AMPK activation. Ex vivo assays showed these effects were likely secondary to blood-borne signals since PBCA sera or conditioned media from PBCA fat progenitors enhanced glucose uptake and activated AMPK in muscle cultures. Thus, adipose progenitor Wnt activation dissociates lipodystrophy from dysfunctional metabolism and highlights a fat-muscle endocrine axis, which may represent a potential therapy to lower blood glucose and improve metabolism.


Subject(s)
Adipocytes/metabolism , Adipocytes/pathology , Glucose/metabolism , Muscles/metabolism , Stem Cells/metabolism , Stem Cells/pathology , Wnt Signaling Pathway , AMP-Activated Protein Kinases/metabolism , Adipocytes/drug effects , Adiposity/drug effects , Animals , Biological Transport/drug effects , Cell Compartmentation/drug effects , Cell Differentiation/drug effects , Cell Lineage/drug effects , Culture Media, Conditioned/pharmacology , Insulin/metabolism , Lipodystrophy/metabolism , Lipodystrophy/pathology , Mice , Mice, Mutant Strains , Muscles/drug effects , Mutation/genetics , PPAR gamma/metabolism , Stem Cells/drug effects , Stromal Cells/drug effects , Stromal Cells/metabolism , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism
14.
Mol Cell Biol ; 31(23): 4692-705, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21947280

ABSTRACT

Osteoclasts are bone-resorbing cells essential for skeletal development, homeostasis, and regeneration. They derive from hematopoietic progenitors in the monocyte/macrophage lineage and differentiate in response to RANKL. However, the precise nature of osteoclast progenitors is a longstanding and important question. Using inducible peroxisome proliferator-activated receptor γ (PPARγ)-tTA TRE-GFP (green fluorescent protein) reporter mice, we show that osteoclast progenitors reside specifically in the PPARγ-expressing hematopoietic bone marrow population and identify the quiescent PPARγ(+) cells as osteoclast progenitors. Importantly, two PPARγ-tTA TRE-Cre-controlled genetic models provide compelling functional evidence. First, Notch activation in PPARγ(+) cells causes high bone mass due to impaired osteoclast precursor proliferation. Second, selective ablation of PPARγ(+) cells by diphtheria toxin also causes high bone mass due to decreased osteoclast numbers. Furthermore, PPARγ(+) cells respond to both pathological and pharmacological resorption-enhancing stimuli. Mechanistically, PPARγ promotes osteoclast progenitors by activating GATA2 transcription. These findings not only identify the long-sought-after osteoclast progenitors but also establish unprecedented tools for their visualization, isolation, characterization, and genetic manipulation.


Subject(s)
Bone Marrow Cells/cytology , GATA2 Transcription Factor/metabolism , Osteoclasts/cytology , PPAR gamma/metabolism , Stem Cells/cytology , Acid Phosphatase/genetics , Acid Phosphatase/metabolism , Animals , Antigens, Differentiation/metabolism , Bone Marrow Cells/metabolism , Cell Differentiation , Cell Lineage , Cells, Cultured , Collagen Type I/urine , Female , GATA2 Transcription Factor/genetics , Gene Expression , Gene Expression Profiling , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Isoenzymes/genetics , Isoenzymes/metabolism , Male , Mice , Mice, Transgenic , Organ Size , Osteocalcin/blood , Osteoclasts/metabolism , PPAR gamma/genetics , Peptide Fragments/urine , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Radiography , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Stem Cells/metabolism , Tartrate-Resistant Acid Phosphatase , Tibia/anatomy & histology , Tibia/cytology , Tibia/diagnostic imaging , Transcription, Genetic
15.
Mol Cell Biol ; 31(23): 4706-19, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21876000

ABSTRACT

Wnt/ß-catenin signaling is a critical regulator of skeletal physiology. However, previous studies have mainly focused on its roles in osteoblasts, while its specific function in osteoclasts is unknown. This is a clinically important question because neutralizing antibodies against Wnt antagonists are promising new drugs for bone diseases. Here, we show that in osteoclastogenesis, ß-catenin is induced during the macrophage colony-stimulating factor (M-CSF)-mediated quiescence-to-proliferation switch but suppressed during the RANKL-mediated proliferation-to-differentiation switch. Genetically, ß-catenin deletion blocks osteoclast precursor proliferation, while ß-catenin constitutive activation sustains proliferation but prevents osteoclast differentiation, both causing osteopetrosis. In contrast, ß-catenin heterozygosity enhances osteoclast differentiation, causing osteoporosis. Biochemically, Wnt activation attenuates whereas Wnt inhibition stimulates osteoclastogenesis. Mechanistically, ß-catenin activation increases GATA2/Evi1 expression but abolishes RANKL-induced c-Jun phosphorylation. Therefore, ß-catenin exerts a pivotal biphasic and dosage-dependent regulation of osteoclastogenesis. Importantly, these findings suggest that Wnt activation is a more effective treatment for skeletal fragility than previously recognized that confers dual anabolic and anti-catabolic benefits.


Subject(s)
Cell Differentiation , Osteoclasts/cytology , beta Catenin/physiology , Animals , Cathepsin K/genetics , Cathepsin K/metabolism , Cells, Cultured , Collagen Type I/blood , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Enzyme Activation , GATA2 Transcription Factor/genetics , GATA2 Transcription Factor/metabolism , Gene Expression , JNK Mitogen-Activated Protein Kinases/metabolism , MDS1 and EVI1 Complex Locus Protein , Macrophage Colony-Stimulating Factor/pharmacology , Macrophage Colony-Stimulating Factor/physiology , Male , Mice , Mice, Transgenic , Osteopetrosis/genetics , Osteopetrosis/metabolism , PPAR gamma/genetics , PPAR gamma/metabolism , Peptide Fragments/blood , Proto-Oncogenes/genetics , RANK Ligand/pharmacology , RANK Ligand/physiology , Radiography , Tibia/anatomy & histology , Tibia/diagnostic imaging , Transcription Factors/genetics , Transcription Factors/metabolism , beta Catenin/genetics , beta Catenin/metabolism
16.
Cell Metab ; 14(1): 116-22, 2011 Jul 06.
Article in English | MEDLINE | ID: mdl-21723509

ABSTRACT

White adipose tissue regulates metabolism; the importance of this control is highlighted by the ongoing pandemic of obesity and associated complications such as diabetes, atherosclerosis, and cancer. White adipose tissue maintenance is a dynamic process, yet very little is known about how pharmacologic stimuli affect such plasticity. Combining in vivo lineage marking and BrdU labeling strategies, we found that rosiglitazone, a member of the thiazolidinedione class of glucose-lowering medicines, markedly increases the evolution of adipose progenitors into adipocytes. Notably, chronic rosiglitazone administration disrupts the adipogenic and self-renewal capacities of the stem cell compartment and alters its molecular characteristics. These data unravel unknown aspects of adipose dynamics and provide a basis to manipulate the adipose lineage for therapeutic ends.


Subject(s)
Adipose Tissue, White/cytology , Thiazolidinediones/pharmacology , Adipocytes/cytology , Adipocytes/metabolism , Adipose Tissue, White/drug effects , Animals , Cell Differentiation , Cell Lineage , Cell Proliferation , Mice , Stem Cells/cytology , Stem Cells/metabolism
17.
Cell Stem Cell ; 5(5): 472-81, 2009 Nov 06.
Article in English | MEDLINE | ID: mdl-19896439

ABSTRACT

We are in the midst of a dire, unprecedented, and global epidemic of obesity and secondary sequelae, most prominently diabetes and hyperlipidemia. Underlying this epidemic is the most hated of cells, adipocytes, and their inherent dynamic ability to expand and renew. This capacity highlights a heretofore undefined stem compartment. Recent in vivo studies, relying upon lineage tracing and flow cytometry methods, have begun to unravel the identity of adipose stem cells, their niche, and the dynamism central to adipose expansion. Thus, the field is moving in a direction that may allow us to manipulate adipose stem cells to beneficial therapeutic ends.


Subject(s)
Adipocytes/cytology , Adult Stem Cells/cytology , Hyperlipidemias/therapy , Lipodystrophy/therapy , Stem Cell Niche , Adipocytes/metabolism , Adipose Tissue/cytology , Adult Stem Cells/metabolism , Animals , Antigens, Differentiation/metabolism , Blood Vessels/cytology , Cell Differentiation , Cell Proliferation , Gene Expression Regulation, Developmental , Hematopoietic Stem Cell Mobilization , Humans , Hyperlipidemias/genetics , Lipodystrophy/genetics , Tissue Transplantation
18.
Science ; 322(5901): 583-6, 2008 Oct 24.
Article in English | MEDLINE | ID: mdl-18801968

ABSTRACT

White adipose (fat) tissues regulate metabolism, reproduction, and life span. Adipocytes form throughout life, with the most marked expansion of the lineage occurring during the postnatal period. Adipocytes develop in coordination with the vasculature, but the identity and location of white adipocyte progenitor cells in vivo are unknown. We used genetically marked mice to isolate proliferating and renewing adipogenic progenitors. We found that most adipocytes descend from a pool of these proliferating progenitors that are already committed, either prenatally or early in postnatal life. These progenitors reside in the mural cell compartment of the adipose vasculature, but not in the vasculature of other tissues. Thus, the adipose vasculature appears to function as a progenitor niche and may provide signals for adipocyte development.


Subject(s)
Adipocytes, White/cytology , Adipose Tissue/blood supply , Blood Vessels/cytology , Multipotent Stem Cells/cytology , Stromal Cells/cytology , Adipocytes, White/metabolism , Adipogenesis , Adipose Tissue/cytology , Animals , Antigens, CD/metabolism , Cell Lineage , Cell Proliferation , Cell Separation , Cells, Cultured , Doxycycline/pharmacology , Gene Expression Profiling , Mice , Mice, Transgenic , Multipotent Stem Cells/metabolism , PPAR gamma/genetics , PPAR gamma/metabolism , Stromal Cells/metabolism
19.
Cell Metab ; 6(3): 195-207, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17767906

ABSTRACT

Adipose (Adp) is an evolutionarily conserved gene isolated from naturally occurring obese flies homozygous for an adp mutation. Here we show that the anti-obesity function of Adp (worm Y73E7A.9, fly adp, and murine Wdtc1) is conserved from worms to mammals. Further, Adp appears to inhibit fat formation in a dosage-sensitive manner. Adp heterozygous flies and Adp heterozygous mutant mice are obese and insulin resistant, as are mice that express a dominant negative form of Adp in fat cells. Conversely, fat-restricted Adp transgenic mice are lean and display improved metabolic profiles. A transient transgenic increase in Adp activity in adult fly fat tissues reduces fat accumulation, indicating therapeutic potential. ADP may elicit these anti-adipogenic functions by regulating chromatin dynamics and gene transcription, as it binds both histones and HDAC3 and inhibits PPARgamma activity. Thus Adp appears to be involved in an ancient pathway that regulates fat accumulation.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/physiology , Obesity/genetics , Proteins/genetics , Proteins/metabolism , Adipose Tissue/anatomy & histology , Adipose Tissue/physiology , Animals , Caenorhabditis elegans/anatomy & histology , Caenorhabditis elegans/physiology , Caenorhabditis elegans Proteins/antagonists & inhibitors , Drosophila Proteins/antagonists & inhibitors , Female , Histone Deacetylases/metabolism , Histones/metabolism , Humans , Hyperglycemia/metabolism , Hyperglycemia/prevention & control , Insulin/metabolism , Male , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Obesity/prevention & control , Proteins/antagonists & inhibitors
20.
Am Psychol ; 62(2): 131-9, 2007.
Article in English | MEDLINE | ID: mdl-17324038

ABSTRACT

In 1974, a story was published about clandestine research done by John B. Watson that was judged to be so reprehensible that it was offered as the real reason he was fired from his faculty position at Johns Hopkins University in 1920, at perhaps the peak of his academic career. Watson's dismissal from Johns Hopkins may have been the most important event in his career, and it almost certainly altered the history of American psychology. Thus, this story has great significance. The claims of the story, however, have never been validated or invalidated. This article examines the evidence for and against the existence of such research and discusses Watson's academic dismissal in light of that evidence.


Subject(s)
Behavioral Research/history , Psychology/history , Sexual Behavior/history , Baltimore , Behavioral Research/methods , Behaviorism/history , History, 20th Century , Humans , Sexual Behavior/physiology , Universities/history
SELECTION OF CITATIONS
SEARCH DETAIL
...