Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
Add more filters










Publication year range
1.
iScience ; 27(5): 109749, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38706850

ABSTRACT

Insulin signaling to the glomerular podocyte via the insulin receptor (IR) is critical for kidney function. In this study we show that near-complete knockout of the closely related insulin-like growth factor 1 receptor (IGF1R) in podocytes is detrimental, resulting in albuminuria in vivo and podocyte cell death in vitro. In contrast, partial podocyte IGF1R knockdown confers protection against doxorubicin-induced podocyte injury. Proteomic analysis of cultured podocytes revealed that while near-complete loss of podocyte IGF1R results in the downregulation of mitochondrial respiratory complex I and DNA damage repair proteins, partial IGF1R inhibition promotes respiratory complex expression. This suggests that altered mitochondrial function and resistance to podocyte stress depends on the level of IGF1R suppression, the latter determining whether receptor inhibition is protective or detrimental. Our work suggests that the partial suppression of podocyte IGF1R could have therapeutic benefits in treating albuminuric kidney disease.

2.
Elife ; 132024 Mar 05.
Article in English | MEDLINE | ID: mdl-38441552

ABSTRACT

The mammary gland is a unique organ that undergoes dynamic alterations throughout a female's reproductive life, making it an ideal model for developmental, stem cell and cancer biology research. Mammary gland development begins in utero and proceeds via a quiescent bud stage before the initial outgrowth and subsequent branching morphogenesis. How mammary epithelial cells transit from quiescence to an actively proliferating and branching tissue during embryogenesis and, importantly, how the branch pattern is determined remain largely unknown. Here, we provide evidence indicating that epithelial cell proliferation and onset of branching are independent processes, yet partially coordinated by the Eda signaling pathway. Through heterotypic and heterochronic epithelial-mesenchymal recombination experiments between mouse mammary and salivary gland tissues and ex vivo live imaging, we demonstrate that unlike previously concluded, the mode of branching is an intrinsic property of the mammary epithelium whereas the pace of growth and the density of ductal tree are determined by the mesenchyme. Transcriptomic profiling and ex vivo and in vivo functional studies in mice disclose that mesenchymal Wnt/ß-catenin signaling, and in particular IGF-1 downstream of it critically regulate mammary gland growth. These results underscore the general need to carefully deconstruct the different developmental processes producing branched organs.


Subject(s)
Epithelial Cells , Wnt Signaling Pathway , Mice , Animals , Epithelium/metabolism , Epithelial Cells/physiology , Cell Proliferation , Morphogenesis , Mesoderm , Mammary Glands, Animal/metabolism
3.
Proc Natl Acad Sci U S A ; 120(25): e2300374120, 2023 06 20.
Article in English | MEDLINE | ID: mdl-37307487

ABSTRACT

When evolution leads to differences in body size, organs generally scale along. A well-known example of the tight relationship between organ and body size is the scaling of mammalian molar teeth. To investigate how teeth scale during development and evolution, we compared molar development from initiation through final size in the mouse and the rat. Whereas the linear dimensions of the rat molars are twice that of the mouse molars, their shapes are largely the same. Here, we focus on the first lower molars that are considered the most reliable dental proxy for size-related patterns due to their low within-species variability. We found that scaling of the molars starts early, and that the rat molar is patterned equally as fast but in a larger size than the mouse molar. Using transcriptomics, we discovered that a known regulator of body size, insulin-like growth factor 1 (Igf1), is more highly expressed in the rat molars compared to the mouse molars. Ex vivo and in vivo mouse models demonstrated that modulation of the IGF pathway reproduces several aspects of the observed scaling process. Furthermore, analysis of IGF1-treated mouse molars and computational modeling indicate that IGF signaling scales teeth by simultaneously enhancing growth and by inhibiting the cusp-patterning program, thereby providing a relatively simple mechanism for scaling teeth during development and evolution. Finally, comparative data from shrews to elephants suggest that this scaling mechanism regulates the minimum tooth size possible, as well as the patterning potential of large teeth.


Subject(s)
Proboscidea Mammal , Rats , Mice , Animals , Molar , Shrews , Body Size , Cognition
4.
Proc Natl Acad Sci U S A ; 119(33): e2121040119, 2022 08 16.
Article in English | MEDLINE | ID: mdl-35943986

ABSTRACT

Regulation of firing rate homeostasis constitutes a fundamental property of central neural circuits. While intracellular Ca2+ has long been hypothesized to be a feedback control signal, the molecular machinery enabling a network-wide homeostatic response remains largely unknown. We show that deletion of insulin-like growth factor-1 receptor (IGF-1R) limits firing rate homeostasis in response to inactivity, without altering the distribution of baseline firing rates. The deficient firing rate homeostatic response was due to disruption of both postsynaptic and intrinsic plasticity. At the cellular level, we detected a fraction of IGF-1Rs in mitochondria, colocalized with the mitochondrial calcium uniporter complex (MCUc). IGF-1R deletion suppressed transcription of the MCUc members and burst-evoked mitochondrial Ca2+ (mitoCa2+) by weakening mitochondria-to-cytosol Ca2+ coupling. Overexpression of either mitochondria-targeted IGF-1R or MCUc in IGF-1R-deficient neurons was sufficient to rescue the deficits in burst-to-mitoCa2+ coupling and firing rate homeostasis. Our findings indicate that mitochondrial IGF-1R is a key regulator of the integrated homeostatic response by tuning the reliability of burst transfer by MCUc. Based on these results, we propose that MCUc acts as a homeostatic Ca2+ sensor. Faulty activation of MCUc may drive dysregulation of firing rate homeostasis in aging and in brain disorders associated with aberrant IGF-1R/MCUc signaling.


Subject(s)
Calcium Channels , Calcium , Receptor, IGF Type 1 , Animals , Calcium/metabolism , Calcium Channels/genetics , Calcium Channels/metabolism , Gene Deletion , Homeostasis , Mice , Neuronal Plasticity , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Reproducibility of Results
5.
Cell Rep ; 39(6): 110793, 2022 05 10.
Article in English | MEDLINE | ID: mdl-35545054

ABSTRACT

Ribosomopathies constitute a range of disorders associated with defective protein synthesis mainly affecting hematopoietic stem cells (HSCs) and erythroid development. Here, we demonstrate that deletion of poly-pyrimidine-tract-binding protein 1 (PTBP1) in the hematopoietic compartment leads to the development of a ribosomopathy-like condition. Specifically, loss of PTBP1 is associated with decreases in HSC self-renewal, erythroid differentiation, and protein synthesis. Consistent with its function as a splicing regulator, PTBP1 deficiency results in splicing defects in hundreds of genes, and we demonstrate that the up-regulation of a specific isoform of CDC42 partly mimics the protein-synthesis defect associated with loss of PTBP1. Furthermore, PTBP1 deficiency is associated with a marked defect in ribosome biogenesis and a selective reduction in the translation of mRNAs encoding ribosomal proteins. Collectively, this work identifies PTBP1 as a key integrator of ribosomal functions and highlights the broad functional repertoire of RNA-binding proteins.


Subject(s)
Hematopoietic Stem Cells , Ribosomes , Erythrocytes/metabolism , Erythropoiesis , Hematopoietic Stem Cells/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Polypyrimidine Tract-Binding Protein/genetics , Polypyrimidine Tract-Binding Protein/metabolism , Ribosomal Proteins/metabolism , Ribosomes/metabolism
6.
Int J Cancer ; 143(10): 2458-2469, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30070361

ABSTRACT

Insulin and insulin-like growth factor (IGF)-1 signaling in the thyroid are thought to be permissive for the coordinated regulation by thyroid-stimulating hormone (TSH) of thyrocyte proliferation and hormone production. However, the integrated role of insulin receptor (IR) and IGF-1 receptor (IGF-1R) in thyroid development and function has not been explored. Here, we generated thyrocyte-specific IR and IGF-1R double knockout (DTIRKO) mice to precisely evaluate the coordinated functions of these receptors in the thyroid of neonates and adults. Neonatal DTIRKO mice displayed smaller thyroids, paralleling defective folliculogenesis associated with repression of the thyroid-specific transcription factor Foxe1. By contrast, at postnatal day 14, absence of IR and IGF-1R paradoxically induced thyrocyte proliferation, which was mediated by mTOR-dependent signaling pathways. Furthermore, we found elevated production of TSH during the development of follicular hyperplasia at 8 weeks of age. By 50 weeks, all DTIRKO mice developed papillary thyroid carcinoma (PTC)-like lesions that correlated with induction of the ErbB pathway. Taken together, these data define a critical role for IR and IGF-1R in neonatal thyroid folliculogenesis. They also reveal an important reciprocal relationship between IR/IGF-1R and TSH/ErbB signaling in the pathogenesis of thyroid follicular hyperplasia and, possibly, of papillary carcinoma.


Subject(s)
ErbB Receptors/metabolism , Receptor, IGF Type 1/deficiency , Receptor, Insulin/deficiency , Thyroid Cancer, Papillary/metabolism , Thyroid Epithelial Cells/metabolism , Thyroid Neoplasms/metabolism , Animals , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Signal Transduction , Thyroid Cancer, Papillary/pathology , Thyroid Epithelial Cells/pathology , Thyroid Neoplasms/pathology , Thyrotropin/biosynthesis , Thyrotropin/metabolism
7.
Curr Biol ; 27(21): 3315-3329.e6, 2017 Nov 06.
Article in English | MEDLINE | ID: mdl-29107547

ABSTRACT

Granule cells (GCs) in the olfactory bulb (OB) play an important role in odor information processing. Although they have been classified into various neurochemical subtypes, the functional roles of these subtypes remain unknown. We used in vivo two-photon Ca2+ imaging combined with cell-type-specific identification of GCs in the mouse OB to examine whether functionally distinct GC subtypes exist in the bulbar network. We showed that half of GCs express Ca2+/calmodulin-dependent protein kinase IIα (CaMKIIα+) and that these neurons are preferentially activated by olfactory stimulation. The higher activity of CaMKIIα+ neurons is due to the weaker inhibitory input that they receive compared to their CaMKIIα-immunonegative (CaMKIIα-) counterparts. In line with these functional data, immunohistochemical analyses showed that 75%-90% of GCs expressing the immediate early gene cFos are CaMKIIα+ in naive animals and in mice that have been exposed to a novel odor and go/no-go operant conditioning, or that have been subjected to long-term associative memory and spontaneous habituation/dishabituation odor discrimination tasks. On the other hand, a perceptual learning task resulted in increased activation of CaMKIIα- cells. Pharmacogenetic inhibition of CaMKIIα+ GCs revealed that this subtype is involved in habituation/dishabituation and go/no-go odor discrimination, but not in perceptual learning. In contrast, pharmacogenetic inhibition of GCs in a subtype-independent manner affected perceptual learning. Our results indicate that functionally distinct populations of GCs exist in the OB and that they play distinct roles during different odor tasks.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Neurons/metabolism , Olfactory Bulb/physiology , Olfactory Perception/physiology , Animals , Behavior, Animal/physiology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/biosynthesis , Male , Mice , Mice, Inbred C57BL , Odorants
8.
Endocrinology ; 158(7): 2269-2283, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28881863

ABSTRACT

Growth hormone (GH) and insulinlike growth factor (IGF) promote aging and age-related pathologies. Inhibiting this pathway by targeting IGF receptor (IGF-1R) is a promising strategy to extend life span, alleviate age-related diseases, and reduce tumor growth. Although anti-IGF-1R agents are being developed, long-term effects of IGF-1R blockade remain unknown. In this study, we used ubiquitous inducible IGF-1R knockout (UBIKOR) to suppress signaling in all adult tissues and screened health extensively. Surprisingly, UBIKOR mice showed no overt defects and presented with rather inconspicuous health, including normal cognition. Endocrine GH and IGF-1 were strongly upregulated without causing acromegaly. UBIKOR mice were strikingly lean with coordinate changes in body composition and organ size. They were insulin resistant but preserved physiological energy expenditure and displayed enhanced fasting metabolic flexibility. Thus, long-term IGF-1R blockade generated beneficial effects on aging-relevant metabolism, but exposed to high GH. This needs to be considered when targeting IGF-1R to protect from neurodegeneration, retard aging, or fight cancer.


Subject(s)
Energy Metabolism/genetics , Growth Hormone/metabolism , Insulin-Like Growth Factor I/metabolism , Receptor, IGF Type 1/genetics , Thinness/genetics , Animals , Body Composition/drug effects , Body Composition/genetics , Energy Metabolism/drug effects , Female , Gene Deletion , Human Growth Hormone/analogs & derivatives , Human Growth Hormone/pharmacology , Insulin Resistance/genetics , Insulin-Like Growth Factor I/antagonists & inhibitors , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptor, IGF Type 1/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/genetics , Thinness/metabolism
9.
Proc Natl Acad Sci U S A ; 114(36): 9731-9736, 2017 09 05.
Article in English | MEDLINE | ID: mdl-28827363

ABSTRACT

When food resources are scarce, endothermic animals can lower core body temperature (Tb). This phenomenon is believed to be part of an adaptive mechanism that may have evolved to conserve energy until more food becomes available. Here, we found in the mouse that the insulin-like growth factor 1 receptor (IGF-1R) controls this response in the central nervous system. Pharmacological or genetic inhibition of IGF-1R enhanced the reduction of temperature and of energy expenditure during calorie restriction. Full blockade of IGF-1R affected female and male mice similarly. In contrast, genetic IGF-1R dosage was effective only in females, where it also induced transient and estrus-specific hypothermia in animals fed ad libitum. These effects were regulated in the brain, as only central, not peripheral, pharmacological activation of IGF-1R prevented hypothermia during calorie restriction. Targeted IGF-1R knockout selectively in forebrain neurons revealed that IGF signaling also modulates calorie restriction-dependent Tb regulation in regions rostral of the canonical hypothalamic nuclei involved in controlling body temperature. In aggregate, these data identify central IGF-1R as a mediator of the integration of nutrient and temperature homeostasis. They also show that calorie restriction, IGF-1R signaling, and body temperature, three of the main regulators of metabolism, aging, and longevity, are components of the same pathway.


Subject(s)
Caloric Restriction/adverse effects , Hypothermia/physiopathology , Receptor, IGF Type 1/physiology , Aging/physiology , Animals , Energy Metabolism/physiology , Female , Gene Dosage , Homeostasis/physiology , Hypothermia/etiology , Hypothermia/prevention & control , Longevity/physiology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Podophyllotoxin/analogs & derivatives , Podophyllotoxin/pharmacology , Receptor, IGF Type 1/antagonists & inhibitors , Receptor, IGF Type 1/genetics , Sex Characteristics , Signal Transduction/physiology
10.
Brain ; 140(7): 2012-2027, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28595357

ABSTRACT

Seminal studies using post-mortem brains of patients with Alzheimer's disease evidenced aberrant insulin-like growth factor 1 receptor (IGF1R) signalling. Addressing causality, work in animal models recently demonstrated that long-term suppression of IGF1R signalling alleviates Alzheimer's disease progression and promotes neuroprotection. However, the underlying mechanisms remain largely elusive. Here, we showed that genetically ablating IGF1R in neurons of the ageing brain efficiently protects from neuroinflammation, anxiety and memory impairments induced by intracerebroventricular injection of amyloid-ß oligomers. In our mutant mice, the suppression of IGF1R signalling also invariably led to small neuronal soma size, indicative of profound changes in cellular homeodynamics. To gain insight into transcriptional signatures leading to Alzheimer's disease-relevant neuronal defence, we performed genome-wide microarray analysis on laser-dissected hippocampal CA1 after neuronal IGF1R knockout, in the presence or absence of APP/PS1 transgenes. Functional analysis comparing neurons in early-stage Alzheimer's disease with IGF1R knockout neurons revealed strongly convergent transcriptomic signatures, notably involving neurite growth, cytoskeleton organization, cellular stress response and neurotransmission. Moreover, in Alzheimer's disease neurons, a high proportion of genes responding to Alzheimer's disease showed a reversed differential expression when IGF1R was deleted. One of the genes consistently highlighted in genome-wide comparison was the neurofilament medium polypeptide Nefm. We found that NEFM accumulated in hippocampus in the presence of amyloid pathology, and decreased to control levels under IGF1R deletion, suggesting that reorganized cytoskeleton likely plays a role in neuroprotection. These findings demonstrated that significant resistance of the brain to amyloid-ß can be achieved lifelong by suppressing neuronal IGF1R and identified IGF-dependent molecular pathways that coordinate an intrinsic program for neuroprotection against proteotoxicity. Our data also indicate that neuronal defences against Alzheimer's disease rely on an endogenous gene expression profile similar to the neuroprotective response activated by genetic disruption of IGF1R signalling. This study highlights neuronal IGF1R signalling as a relevant target for developing Alzheimer's disease prevention strategies.


Subject(s)
Alzheimer Disease/metabolism , CA1 Region, Hippocampal/metabolism , Neuroprotective Agents/metabolism , Receptor, IGF Type 1/deficiency , Receptor, IGF Type 1/genetics , Transcriptome , Aging/metabolism , Alzheimer Disease/complications , Amyloid beta-Peptides/administration & dosage , Animals , Anxiety/chemically induced , Anxiety/complications , Anxiety/prevention & control , Encephalitis/chemically induced , Encephalitis/complications , Encephalitis/prevention & control , Female , Infusions, Intraventricular , Male , Memory Disorders/chemically induced , Memory Disorders/complications , Memory Disorders/prevention & control , Mice , Mice, Knockout , Mice, Transgenic , Neurons/metabolism
11.
Exp Dermatol ; 26(9): 785-791, 2017 09.
Article in English | MEDLINE | ID: mdl-28094870

ABSTRACT

Insulin-like growth factor 1 (Igf1) is important for skin development and homoeostasis. However, overexpression and inactivation studies have produced variable findings regarding its role in hair follicle (HF) biology. Here, we studied a conditional and inducible knockout of the Igf1 receptor (Igf1r) in keratin 15-expressing bulge cells. Deletion of Igf1r after the development of the skin appendages in K15-Igf1rKO mice showed no abnormalities in epidermal homoeostasis. Numbers of bulge cells were lower in K15-Igf1rKO mice than in controls, without consequences on wound healing, at least in young mice. K15-Igf1rKO HFs entered anagen phase earlier than controls and showed a delay in the anagen/catagen switch. The expression of Bmp-4 mRNA was inhibited in HFs from K15-Igf1rKO . MED1 transcription was impaired in the epidermis of K15-Igf1rKO mice. These findings suggest that Igf1r controls the hair cycle, partly through Bmp-4 activation.


Subject(s)
Hair/growth & development , Insulin-Like Growth Factor I/metabolism , Receptor, IGF Type 1/metabolism , Animals , Epidermis/physiology , Male , Mice, Knockout , Receptor, IGF Type 1/genetics
12.
J Cereb Blood Flow Metab ; 37(2): 396-412, 2017 Feb.
Article in English | MEDLINE | ID: mdl-26762506

ABSTRACT

Insulin-like growth factors control numerous processes, namely somatic growth, metabolism and stress resistance, connecting this pathway to aging and age-related diseases. Insulin-like growth factor signaling also impacts on neurogenesis, neuronal survival and structural plasticity. Recent reports demonstrated that diminished insulin-like growth factor signaling confers increased stress resistance in brain and other tissues. To better understand the role of neuronal insulin-like growth factor signaling in neuroprotection, we inactivated insulin-like growth factor type-1-receptor in forebrain neurons using conditional Cre-LoxP-mediated gene targeting. We found that brain structure and function, including memory performance, were preserved in insulin-like growth factor receptor mutants, and that certain characteristics improved, notably synaptic transmission in hippocampal neurons. To reveal stress-related roles of insulin-like growth factor signaling, we challenged the brain using a stroke-like insult. Importantly, when charged with hypoxia-ischemia, mutant brains were broadly protected from cell damage, neuroinflammation and cerebral edema. We also found that in mice with insulin-like growth factor receptor knockout specifically in forebrain neurons, a substantial systemic upregulation of growth hormone and insulin-like growth factor-I occurred, which was associated with significant somatic overgrowth. Collectively, we found strong evidence that blocking neuronal insulin-like growth factor signaling increases peripheral somatotropic tone and simultaneously protects the brain against hypoxic-ischemic injury, findings that may contribute to developing new therapeutic concepts preventing the disabling consequences of stroke.


Subject(s)
Gene Deletion , Growth Hormone/metabolism , Neuroprotection , Prosencephalon/pathology , Receptor, IGF Type 1/genetics , Stroke/genetics , Stroke/pathology , Animals , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Neurons/pathology , Prosencephalon/metabolism , Stroke/metabolism , Up-Regulation
13.
Neurobiol Aging ; 41: 64-72, 2016 May.
Article in English | MEDLINE | ID: mdl-27103519

ABSTRACT

Hypothalamic tanycytes are specialized glial cells lining the third ventricle. They are recently identified as adult stem and/or progenitor cells, able to self-renew and give rise to new neurons postnatally. However, the long-term neurogenic potential of tanycytes and the pathways regulating lifelong cell replacement in the adult hypothalamus are largely unexplored. Using inducible nestin-CreER(T2) for conditional mutagenesis, we performed lineage tracing of adult hypothalamic stem and/or progenitor cells (HySC) and demonstrated that new neurons continue to be born throughout adult life. This neurogenesis was targeted to numerous hypothalamic nuclei and produced different types of neurons in the dorsal periventricular regions. Some adult-born neurons integrated the median eminence and arcuate nucleus during aging and produced growth hormone releasing hormone. We showed that adult hypothalamic neurogenesis was tightly controlled by insulin-like growth factors (IGF). Knockout of IGF-1 receptor from hypothalamic stem and/or progenitor cells increased neuronal production and enhanced α-tanycyte self-renewal, preserving this stem cell-like population from age-related attrition. Our data indicate that adult hypothalamus retains the capacity of cell renewal, and thus, a substantial degree of structural plasticity throughout lifespan.


Subject(s)
Aging/physiology , Hypothalamus/cytology , Hypothalamus/growth & development , Insulin-Like Growth Factor I/physiology , Neurogenesis/genetics , Neurogenesis/physiology , Signal Transduction/genetics , Signal Transduction/physiology , Aging/pathology , Animals , Cell Plasticity , Cell Self Renewal , Ependymoglial Cells/cytology , Male , Mice, Transgenic , Models, Animal
14.
Brain ; 139(Pt 4): 1237-51, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26912648

ABSTRACT

Recent studies highlight the implication of innate and adaptive immunity in the pathophysiology of Alzheimer's disease, and foster immunotherapy as a promising strategy for its treatment. Vaccines targeting amyloid-ß peptide provided encouraging results in mouse models, but severe side effects attributed to T cell responses in the first clinical trial AN1792 underlined the need for better understanding adaptive immunity in Alzheimer's disease. We previously showed that regulatory T cells critically control amyloid-ß-specific CD4(+) T cell responses in both physiological and pathological settings. Here, we analysed the impact of regulatory T cells on spontaneous disease progression in a murine model of Alzheimer's disease. Early transient depletion of regulatory T cells accelerated the onset of cognitive deficits in APPPS1 mice, without altering amyloid-ß deposition. Earlier cognitive impairment correlated with reduced recruitment of microglia towards amyloid deposits and altered disease-related gene expression profile. Conversely, amplification of regulatory T cells through peripheral low-dose IL-2 treatment increased numbers of plaque-associated microglia, and restored cognitive functions in APPPS1 mice. These data suggest that regulatory T cells play a beneficial role in the pathophysiology of Alzheimer's disease, by slowing disease progression and modulating microglial response to amyloid-ß deposition. Our study highlights the therapeutic potential of repurposed IL-2 for innovative immunotherapy based on modulation of regulatory T cells in Alzheimer's disease.


Subject(s)
Alzheimer Disease/immunology , Alzheimer Disease/pathology , Disease Progression , T-Lymphocytes, Regulatory/physiology , Alzheimer Disease/prevention & control , Amyloid beta-Protein Precursor , Animals , Brain/immunology , Brain/pathology , Humans , Interleukin-2/pharmacology , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Presenilin-1 , T-Lymphocytes, Regulatory/drug effects
15.
J Neurosci ; 35(34): 12063-79, 2015 Aug 26.
Article in English | MEDLINE | ID: mdl-26311784

ABSTRACT

Spinal muscular atrophy (SMA) is a neuromuscular disease characterized by the selective loss of spinal motor neurons due to the depletion of the survival of motor neuron (SMN) protein. No therapy is currently available for SMA, which represents the leading genetic cause of death in childhood. In the present study, we report that insulin-like growth factor-1 receptor (Igf-1r) gene expression is enhanced in the spinal cords of SMA-like mice. The reduction of expression, either at the physiological (through physical exercise) or genetic level, resulted in the following: (1) a significant improvement in lifespan and motor behavior, (2) a significant motor neuron protection, and (3) an increase in SMN expression in spinal cord and skeletal muscles through both transcriptional and posttranscriptional mechanisms. Furthermore, we have found that reducing IGF-1R expression is sufficient to restore intracellular signaling pathway activation profile lying downstream of IGF-1R, resulting in both the powerful activation of the neuroprotective AKT/CREB pathway and the inhibition of the ERK and JAK pathways. Therefore, reducing rather than enhancing the IGF-1 pathway could constitute a useful strategy to limit neurodegeneration in SMA. SIGNIFICANCE STATEMENT: Recent evidence of IGF-1 axis alteration in spinal muscular atrophy (SMA), a very severe neurodegenerative disease affecting specifically the motor neurons, have triggered a renewed interest in insulin-like growth factor-1 (IGF-1) pathway activation as a potential therapeutic approach for motor neuron diseases. The present study challenges this point of view and brings the alternative hypothesis that reducing rather than enhancing the IGF-1 signaling pathway exerts a neuroprotective effect in SMA. Furthermore, the present data substantiate a newly emerging concept that the modulation of IGF-1 receptor expression is a key event selectively determining the activation level of intracellular pathways that lie downstream of the receptor. This aspect should be considered when designing IGF-1-based treatments for neurodegenerative diseases.


Subject(s)
Muscular Atrophy, Spinal/metabolism , Muscular Atrophy, Spinal/prevention & control , Receptor, IGF Type 1/metabolism , Signal Transduction/physiology , Animals , Cells, Cultured , Female , Humans , Male , Mice , Mice, Knockout , Mice, Transgenic , Muscular Atrophy, Spinal/genetics , Receptor, IGF Type 1/genetics
16.
J Neurosci ; 35(33): 11500-13, 2015 Aug 19.
Article in English | MEDLINE | ID: mdl-26290229

ABSTRACT

Alzheimer's disease (AD) is a frequent and irreversible age-related neurodegeneration without efficient treatment. Experimental AD in mice responds positively to decreased insulin-like growth factor I (IGF-I) signaling, a pathway also implicated in aging. Here we aimed to protect the aging brain from devastating amyloid pathology by making specifically adult neurons resistant to IGF signaling. To achieve that, we knocked out neuronal IGF-1R during adulthood in APP/PS1 mice. We found that mutants exhibited improved spatial memory and reduced anxiety. Mutant brains displayed fewer amyloid plaques, less amyloid-ß (Aß), and diminished neuroinflammation. Surprisingly, adult neurons undergoing IGF-1R knock-out reduced their apical soma and developed leaner dendrites, indicative of remarkable structural plasticity entailing condensed forebrain neuroarchitecture. Neurons lacking IGF-1R in AD showed less accumulation of Aß-containing autophagic vacuoles. At the same time, plasma Aß levels were increased. Our data indicate that neuronal IGF-1R ablation, via preserved autophagic compartment and enhanced systemic elimination, offers lifelong protection from AD pathology by clearing toxic Aß. Neuronal IGF-1R, and possibly other cell size-controlling pathways are promising targets for AD treatment. SIGNIFICANCE STATEMENT: We found compelling evidence in vivo that Alzheimer's disease (AD) progression is significantly delayed when insulin-like growth factor (IGF) signaling is blocked in adult neurons. To show that, we built a novel mouse model, combining inducible neuron-specific IGF-1R knock-out with AD transgenics. Analysis of the experimental AD phenotype revealed less abundant amyloid-ß (Aß) peptides, fewer plaques, and diminished neuroinflammation in mutants with inactivated IGF signaling, together with clearly preserved behavioral and memory performances. We present for the first time evidence that IGF signaling has profound effects on neuronal proteostasis and maintenance of cell morphology in vivo. Our results indicate in a model highly pertinent to translational research that neuronal IGF resistance may represent a pathophysiologically relevant mechanism of the brain for preventing Aß accumulation.


Subject(s)
Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Insulin-Like Growth Factor I/metabolism , Neurons/metabolism , Neurons/pathology , Animals , Behavior, Animal , Cell Size , Cells, Cultured , Down-Regulation , Female , Male , Maze Learning , Metabolic Clearance Rate , Mice , Mice, Knockout , Mice, Transgenic , Prosencephalon/metabolism , Prosencephalon/pathology , Receptor, IGF Type 1/genetics , Signal Transduction
17.
Aging Cell ; 14(5): 847-56, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26219530

ABSTRACT

Downregulation of insulin-like growth factor (IGF) pathways prolongs lifespan in various species, including mammals. Still, the cellular mechanisms by which IGF signaling controls the aging trajectory of individual organs are largely unknown. Here, we asked whether suppression of IGF-I receptor (IGF-1R) in adult stem cells preserves long-term cell replacement, and whether this may prevent age-related functional decline in a regenerating tissue. Using neurogenesis as a paradigm, we showed that conditional knockout of IGF-1R specifically in adult neural stem cells (NSC) maintained youthful characteristics of olfactory bulb neurogenesis within an aging brain. We found that blocking IGF-I signaling in neural precursors increased cumulative neuroblast production and enhanced neuronal integration into the olfactory bulb. This in turn resulted in neuro-anatomical changes that improved olfactory function. Interestingly, mutants also displayed long-term alterations in energy metabolism, possibly related to IGF-1R deletion in NSCs throughout lifespan. We explored Akt and ERK signaling cascades and revealed differential regulation downstream of IGF-1R, with Akt phosphorylation preferentially decreased in IGF-1R(-/-) NSCs within the niche, and ERK pathway downregulated in differentiated neurons of the OB. These challenging experimental results were sustained by data from mathematical modeling, predicting that diminished stimulation of growth is indeed optimal for tissue aging. Thus, inhibiting growth and longevity gene IGF-1R in adult NSCs induced a gain-of-function phenotype during aging, marked by optimized management of cell renewal, and enhanced olfactory sensory function.


Subject(s)
Aging/metabolism , Neural Stem Cells/metabolism , Neurogenesis , Olfactory Bulb/metabolism , Receptor, IGF Type 1/metabolism , Signal Transduction , Animals , Mice , Mice, Knockout , Receptor, IGF Type 1/deficiency , Receptor, IGF Type 1/genetics
18.
PLoS One ; 10(3): e0119392, 2015.
Article in English | MEDLINE | ID: mdl-25793522

ABSTRACT

BACKGROUND: In the testis, thyroid hormone (T3) regulates the number of gametes produced through its action on Sertoli cell proliferation. However, the role of T3 in the regulation of steroidogenesis is still controversial. METHODS: The TRαAMI knock-in allele allows the generation of transgenic mice expressing a dominant-negative TRα1 (thyroid receptor α1) isoform restricted to specific target cells after Cre-loxP recombination. Here, we introduced this mutant allele in both Sertoli and Leydig cells using a novel aromatase-iCre (ARO-iCre) line that expresses Cre recombinase under control of the human Cyp19(IIa)/aromatase promoter. FINDINGS: We showed that loxP recombination induced by this ARO-iCre is restricted to male and female gonads, and is effective in Sertoli and Leydig cells, but not in germ cells. We compared this model with the previous introduction of TRαAMI specifically in Sertoli cells in order to investigate T3 regulation of steroidogenesis. We demonstrated that TRαAMI-ARO males exhibited increased testis weight, increased sperm reserve in adulthood correlated to an increased proliferative index at P3 in vivo, and a loss of T3-response in vitro. Nevertheless, TRαAMI-ARO males showed normal fertility. This phenotype is similar to TRαAMI-SC males. Importantly, plasma testosterone and luteinizing hormone levels, as well as mRNA levels of steroidogenesis enzymes StAR, Cyp11a1 and Cyp17a1 were not affected in TRαAMI-ARO. CONCLUSIONS/SIGNIFICANCE: We concluded that the presence of a mutant TRαAMI allele in both Leydig and Sertoli cells does not accentuate the phenotype in comparison with its presence in Sertoli cells only. This suggests that direct T3 regulation of steroidogenesis through TRα1 is moderate in Leydig cells, and that Sertoli cells are the main target of T3 action in the testis.


Subject(s)
Gene Expression , Leydig Cells/metabolism , Sertoli Cells/metabolism , Thyroid Hormone Receptors alpha/genetics , Animals , Cell Proliferation , Fertility , Gene Targeting , Humans , Luteinizing Hormone/blood , Male , Mice , Mice, Transgenic , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spermatozoa/metabolism , Testis/metabolism , Testosterone/blood
19.
J Hematol Oncol ; 8: 3, 2015 Feb 04.
Article in English | MEDLINE | ID: mdl-25648584

ABSTRACT

BACKGROUND: The tyrosine kinase receptor insulin-like growth factor 1 receptor (IGF-IR) contributes to the initiation and progression of many types of malignancies. We previously showed that IGF-2, which binds IGF-IR, is an extrinsic factor that supports the ex vivo expansion of hematopoietic stem cells (HSCs). We also demonstrated that IGF-IR is not required for HSC activity in vivo. METHODS AND RESULTS: Here we investigated the role of IGF-IR in chronic myeloid leukemia (CML) using the retroviral BCR/ABL transplantation mouse model. Existing antibodies against IGF-IR are not suitable for flow cytometry; therefore, we generated a fusion of the human IgG Fc fragment with mutant IGF-2 that can bind to IGF-IR. We used this fusion protein to evaluate mouse primary hematopoietic populations. Through transplantation assays with IGF-IR(+) and IGF-IR(-) cells, we demonstrated that IGF-IR is expressed on all mouse HSCs. The expression of IGF-IR is much higher on CML cells than on acute lymphoblastic leukemia (ALL) cells. The depletion of IGF-IR expression in BCR/ABL(+) cells led to the development of ALL (mostly T cell ALL) but not CML. Lack of IGF-IR resulted in decreased self-renewal of the BCR/ABL(+) CML cells in the serial replating assay. CONCLUSION: IGF-IR regulates the cell fate determination of BCR/ABL(+) leukemia cells and supports the self-renewal of CML cells.


Subject(s)
Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Receptor, IGF Type 1/metabolism , Animals , Blotting, Western , Flow Cytometry , Fusion Proteins, bcr-abl/genetics , Humans , Immunohistochemistry , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Mice , Mice, Inbred C57BL , Reverse Transcriptase Polymerase Chain Reaction , Transfection
20.
Endocrinology ; 155(2): 370-9, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24265452

ABSTRACT

Glucagon-like peptide-2 (GLP-2) is an intestinal growth-promoting hormone used to treat short bowel syndrome. GLP-2 promotes intestinal growth through a mechanism that involves both IGF-1 and the intestinal-epithelial IGF-1 receptor (IE-IGF-1R). GLP-2 also enhances intestinal barrier function, but through an unknown mechanism. We therefore hypothesized that GLP-2-enhanced barrier function requires the IE-IGF-1R and is mediated through alterations in expression and localization of tight junction proteins. Conditional IE-IGF-1R-null and control mice were treated with vehicle or degradation-resistant Gly(2)-GLP-2 for 10 days; some animals also received irinotecan to induce enteritis. Mice were then examined for gastrointestinal permeability to 4-kDa fluorescein isothiocyanate-dextran, jejunal resistance using Ussing chambers, tight junction structure by electron microscopy, and expression and localization of tight junction proteins by immunoblot and immunohistofluorescence, respectively. GLP-2 treatment decreased permeability to 4-kDa fluorescein isothiocyanate-dextran and increased jejunal resistance (P <.05-.01), effects that were lost in IE-IGF-1R-null mice. Electron microscopy did not reveal major structural changes in the tight junctions in any group of animals. However, the tight junctional proteins claudin-3 and -7 were upregulated by GLP-2 in control (P <.05-.01) but not null mice, whereas IE-IGF-1R deletion induced a shift in occludin localization from apical to intracellular domains; no changes were observed in expression or distribution of claudin-15 and zona occludins-1. Finally, in irinotecan-induced enteritis, GLP-2 normalized epithelial barrier function in control (P < .05) but not knockout animals. In conclusion, the effects of GLP-2 on intestinal barrier function are dependent on the IE-IGF-1R and involve modulation of key components of the tight junctional complex.


Subject(s)
Enteritis/drug therapy , Glucagon-Like Peptide 2/pharmacology , Intestinal Mucosa/metabolism , Receptor, IGF Type 1/metabolism , Animals , Claudin-3/metabolism , Claudins/metabolism , Enteritis/chemically induced , Enteritis/metabolism , Glucagon-Like Peptide 2/therapeutic use , Intestinal Mucosa/drug effects , Intestines/drug effects , Mice , Mice, Knockout , Permeability/drug effects , Receptor, IGF Type 1/genetics , Tight Junctions/metabolism , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL