Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 27
1.
JHEP Rep ; 5(2): 100615, 2023 Feb.
Article En | MEDLINE | ID: mdl-36687468

Background & Aims: Histological assessment of liver biopsies is the gold standard for diagnosis of non-alcoholic steatohepatitis (NASH), the progressive form of non-alcoholic fatty liver disease (NAFLD), despite its well-established limitations. Therefore, non-invasive biomarkers that can offer an integrated view of the liver are needed to improve diagnosis and reduce sampling bias. Hepatic stellate cells (HSCs) are central in the development of hepatic fibrosis, a hallmark of NASH. Secreted HSC-specific proteins may, therefore, reflect disease state in the NASH liver and serve as non-invasive diagnostic biomarkers. Methods: We performed RNA-sequencing on liver biopsies from a histologically characterised cohort of obese patients (n = 30, BMI >35 kg/m2) to identify and evaluate HSC-specific genes encoding secreted proteins. Bioinformatics was used to identify potential biomarkers and their expression at single-cell resolution. We validated our findings using single-molecule fluorescence in situ hybridisation (smFISH) and ELISA to detect mRNA in liver tissue and protein levels in plasma, respectively. Results: Hepatic expression of SPARC-related modular calcium-binding protein 2 (SMOC2) was increased in NASH compared to no-NAFLD (p.adj <0.001). Single-cell RNA-sequencing data indicated that SMOC2 was primarily expressed by HSCs, which was validated using smFISH. Finally, plasma SMOC2 was elevated in NASH compared to no-NAFLD (p <0.001), with a predictive accuracy of AUROC 0.88. Conclusions: Increased SMOC2 in plasma appears to reflect HSC activation, a key cellular event associated with NASH progression, and may serve as a non-invasive biomarker of NASH. Impact and implications: Non-alcoholic fatty liver disease (NAFLD) and its progressive form, non-alcoholic steatohepatitis (NASH), are the most common forms of chronic liver diseases. Currently, liver biopsies are the gold standard for diagnosing NAFLD. Blood-based biomarkers to complement liver biopsies for diagnosis of NAFLD are required. We found that activated hepatic stellate cells, a cell type central to NAFLD pathogenesis, upregulate expression of the secreted protein SPARC-related modular calcium-binding protein 2 (SMOC2). SMOC2 was elevated in blood samples from patients with NASH and may hold promise as a blood-based biomarker for the diagnosis of NAFLD.

2.
Cells ; 10(11)2021 10 20.
Article En | MEDLINE | ID: mdl-34831029

Background: Although several approaches have revealed much about individual factors that regulate pancreatic development, we have yet to fully understand their complicated interplay during pancreas morphogenesis. Gfi1 is transcription factor specifically expressed in pancreatic acinar cells, whose role in pancreas cells fate identity and specification is still elusive. Methods: In order to gain further insight into the function of this factor in the pancreas, we generated animals deficient for Gfi1 specifically in the pancreas. Gfi1 conditional knockout animals were phenotypically characterized by immunohistochemistry, RT-qPCR, and RNA scope. To assess the role of Gfi1 in the pathogenesis of diabetes, we challenged Gfi1-deficient mice with two models of induced hyperglycemia: long-term high-fat/high-sugar feeding and streptozotocin injections. Results: Interestingly, mutant mice did not show any obvious deleterious phenotype. However, in depth analyses demonstrated a significant decrease in pancreatic amylase expression, leading to a diminution in intestinal carbohydrates processing and thus glucose absorption. In fact, Gfi1-deficient mice were found resistant to diet-induced hyperglycemia, appearing normoglycemic even after long-term high-fat/high-sugar diet. Another feature observed in mutant acinar cells was the misexpression of ghrelin, a hormone previously suggested to exhibit anti-apoptotic effects on ß-cells in vitro. Impressively, Gfi1 mutant mice were found to be resistant to the cytotoxic and diabetogenic effects of high-dose streptozotocin administrations, displaying a negligible loss of ß-cells and an imperturbable normoglycemia. Conclusions: Together, these results demonstrate that Gfi1 could turn to be extremely valuable for the development of new therapies and could thus open new research avenues in the context of diabetes research.


DNA-Binding Proteins/deficiency , Diabetes Mellitus/metabolism , Diabetes Mellitus/prevention & control , Transcription Factors/deficiency , Acinar Cells/cytology , Acinar Cells/metabolism , Amylases/metabolism , Animals , Cell Differentiation/genetics , Cell Proliferation/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Diabetes Mellitus/genetics , Disease Models, Animal , Gene Expression Regulation , Ghrelin/metabolism , Homeodomain Proteins/metabolism , Hyperglycemia/complications , Hyperglycemia/genetics , Integrases/metabolism , Mice, Transgenic , Mutation/genetics , Pancreas/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
3.
Sci Rep ; 10(1): 14052, 2020 08 20.
Article En | MEDLINE | ID: mdl-32820201

C57BL/6J-related mouse strains are widely used animal models for diet-induced obesity (DIO). Multiple vendors breed C57BL/6J-related substrains which may introduce genetic drift and environmental confounders such as microbiome differences. To address potential vendor/substrain specific effects, we compared DIO of C57BL/6J-related substrains from three different vendors: C57BL/6J (Charles Rivers), C57BL/6JBomTac (Taconic Bioscience) and C57BL/6JRj (Janvier). After local acclimatization, DIO was induced by either a high-fat diet (HFD, 60% energy from fat) or western diet (WD, 42% energy from fat supplemented with fructose in the drinking water). All three groups on HFD gained a similar amount of total body weight, yet the relative amount of fat percentage and mass of inguinal- and epididymal white adipose tissue (iWAT and eWAT) was lower in C57BL/6JBomTac compared to the two other C57BL/6J-releated substrains. In contrast to HFD, the three groups on WD responded differently in terms of body weight gain, where C57BL/6J was particularly prone to WD. This was associated with a relative higher amount of eWAT, iWAT, and liver triglycerides. Although the HFD and WD had significant impact on the microbiota, we did not observe any major differences between the three groups of mice. Together, these data demonstrate significant differences in HFD- and WD-induced adiposity in C57BL/6J-related substrains, which should be considered in the design of animal DIO studies.


Diet, High-Fat , Absorptiometry, Photon , Animals , Body Weight , Glucose/administration & dosage , Insulin/blood , Liver/metabolism , Mice , Mice, Inbred C57BL , Obesity/genetics , Organ Size , Species Specificity , Triglycerides/metabolism , Weight Gain
4.
PLoS One ; 13(8): e0201536, 2018.
Article En | MEDLINE | ID: mdl-30092080

In the context of type 1 diabetes research and the development of insulin-producing ß-cell replacement strategies, whether pancreatic ductal cells retain their developmental capability to adopt an endocrine cell identity remains debated, most likely due to the diversity of models employed to induce pancreatic regeneration. In this work, rather than injuring the pancreas, we developed a mouse model allowing the inducible misexpression of the proendocrine gene Neurog3 in ductal cells in vivo. These animals developed a progressive islet hypertrophy attributed to a proportional increase in all endocrine cell populations. Lineage tracing experiments indicated a continuous neo-generation of endocrine cells exhibiting a ductal ontogeny. Interestingly, the resulting supplementary ß-like cells were found to be functional. Based on these findings, we suggest that ductal cells could represent a renewable source of new ß-like cells and that strategies aiming at controlling the expression of Neurog3, or of its molecular targets/co-factors, may pave new avenues for the improved treatments of diabetes.


Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Plasticity/physiology , Diabetes Mellitus, Type 1/pathology , Endocrine Cells/physiology , Nerve Tissue Proteins/metabolism , Pancreatic Ducts/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Diabetes Mellitus, Type 1/genetics , Disease Models, Animal , Humans , Insulin-Secreting Cells/metabolism , Male , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Pancreatic Ducts/cytology , Regeneration
6.
J Cell Biol ; 216(12): 4299-4311, 2017 12 04.
Article En | MEDLINE | ID: mdl-29025873

The recent demonstration that pancreatic α cells can be continuously regenerated and converted into ß-like cells upon ectopic expression of Pax4 opened new avenues of research in the endocrine cell differentiation and diabetes fields. To determine whether such plasticity was also shared by δ cells, we generated and characterized transgenic animals that express Pax4 specifically in somatostatin-expressing cells. We demonstrate that the ectopic expression of Pax4 in δ cells is sufficient to induce their conversion into functional ß-like cells. Importantly, this conversion induces compensatory mechanisms involving the reactivation of endocrine developmental processes that result in dramatic ß-like cell hyperplasia. Importantly, these ß-like cells are functional and can partly reverse the consequences of chemically induced diabetes.


Diabetes Mellitus, Experimental/genetics , Ectopic Gene Expression , Homeodomain Proteins/genetics , Insulin-Secreting Cells/metabolism , Paired Box Transcription Factors/genetics , Somatostatin-Secreting Cells/metabolism , Animals , Cell Proliferation , Cell Transdifferentiation/genetics , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/therapy , Genetic Therapy/methods , Glucagon/biosynthesis , Glucagon/genetics , Homeodomain Proteins/metabolism , Insulin/biosynthesis , Insulin/genetics , Insulin-Secreting Cells/cytology , Male , Mice , Mice, Transgenic , Paired Box Transcription Factors/metabolism , Somatostatin/biosynthesis , Somatostatin/genetics , Somatostatin-Secreting Cells/cytology , Streptozocin
7.
Commun Integr Biol ; 10(3): e1300215, 2017.
Article En | MEDLINE | ID: mdl-28702122

Diabetes is a chronic and progressing disease, the number of patients increasing exponentially, especially in industrialized countries. Regenerating lost insulin-producing cells would represent a promising therapeutic alternative for most diabetic patients. To this end, using the mouse as a model, we reported that GABA, a food supplement, could induce insulin-producing beta-like cell neogenesis offering an attractive and innovative approach for diabetes therapeutics.

8.
Front Genet ; 8: 75, 2017.
Article En | MEDLINE | ID: mdl-28634486

Type 1 diabetes is an auto-immune disease resulting in the loss of pancreatic ß-cells and, consequently, in chronic hyperglycemia. Insulin supplementation allows diabetic patients to control their glycaemia quite efficiently, but treated patients still display an overall shortened life expectancy and an altered quality of life as compared to their healthy counterparts. In this context and due to the ever increasing number of diabetics, establishing alternative therapies has become a crucial research goal. Most current efforts therefore aim at generating fully functional insulin-secreting ß-like cells using multiple approaches. In this review, we screened the literature published since 2011 and inventoried the selected markers used to characterize insulin-secreting cells generated by in vitro differentiation of stem/precursor cells or by means of in vivo transdifferentiation. By listing these features, we noted important discrepancies when comparing the different approaches for the initial characterization of insulin-producing cells as true ß-cells. Considering the recent advances achieved in this field of research, the necessity to establish strict guidelines has become a subject of crucial importance, especially should one contemplate the next step, which is the transplantation of in vitro or ex vivo generated insulin-secreting cells in type 1 diabetic patients.

9.
Cell ; 168(1-2): 73-85.e11, 2017 Jan 12.
Article En | MEDLINE | ID: mdl-27916274

The recent discovery that genetically modified α cells can regenerate and convert into ß-like cells in vivo holds great promise for diabetes research. However, to eventually translate these findings to human, it is crucial to discover compounds with similar activities. Herein, we report the identification of GABA as an inducer of α-to-ß-like cell conversion in vivo. This conversion induces α cell replacement mechanisms through the mobilization of duct-lining precursor cells that adopt an α cell identity prior to being converted into ß-like cells, solely upon sustained GABA exposure. Importantly, these neo-generated ß-like cells are functional and can repeatedly reverse chemically induced diabetes in vivo. Similarly, the treatment of transplanted human islets with GABA results in a loss of α cells and a concomitant increase in ß-like cell counts, suggestive of α-to-ß-like cell conversion processes also in humans. This newly discovered GABA-induced α cell-mediated ß-like cell neogenesis could therefore represent an unprecedented hope toward improved therapies for diabetes.


Diabetes Mellitus/drug therapy , Glucagon-Secreting Cells/cytology , Insulin-Secreting Cells/cytology , gamma-Aminobutyric Acid/administration & dosage , Animals , Basic Helix-Loop-Helix Transcription Factors , Cell Differentiation/drug effects , Diabetes Mellitus/chemically induced , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Glucagon-Secreting Cells/drug effects , Humans , Islets of Langerhans/cytology , Male , Mice , Nerve Tissue Proteins , Rats , Rats, Wistar , gamma-Aminobutyric Acid/pharmacology
10.
Cell ; 168(1-2): 86-100.e15, 2017 Jan 12.
Article En | MEDLINE | ID: mdl-27916275

Type 1 diabetes is characterized by the destruction of pancreatic ß cells, and generating new insulin-producing cells from other cell types is a major aim of regenerative medicine. One promising approach is transdifferentiation of developmentally related pancreatic cell types, including glucagon-producing α cells. In a genetic model, loss of the master regulatory transcription factor Arx is sufficient to induce the conversion of α cells to functional ß-like cells. Here, we identify artemisinins as small molecules that functionally repress Arx by causing its translocation to the cytoplasm. We show that the protein gephyrin is the mammalian target of these antimalarial drugs and that the mechanism of action of these molecules depends on the enhancement of GABAA receptor signaling. Our results in zebrafish, rodents, and primary human pancreatic islets identify gephyrin as a druggable target for the regeneration of pancreatic ß cell mass from α cells.


Artemisinins/pharmacology , Diabetes Mellitus, Type 1/drug therapy , Disease Models, Animal , Receptors, GABA-A/metabolism , Signal Transduction , Animals , Artemether , Artemisinins/administration & dosage , Carrier Proteins/metabolism , Cell Transdifferentiation/drug effects , Cells, Cultured , Diabetes Mellitus/drug therapy , Diabetes Mellitus, Type 1/pathology , Gene Expression Profiling , Homeodomain Proteins/metabolism , Humans , Insulin/genetics , Insulin/metabolism , Islets of Langerhans/drug effects , Membrane Proteins/metabolism , Mice , Protein Stability/drug effects , Rats , Single-Cell Analysis , Transcription Factors/metabolism , Zebrafish , gamma-Aminobutyric Acid/metabolism
11.
PLoS One ; 10(9): e0138668, 2015.
Article En | MEDLINE | ID: mdl-26405811

We have previously reported an antisense technology, 'snoMEN vectors', for targeted knock-down of protein coding mRNAs using human snoRNAs manipulated to contain short regions of sequence complementarity with the mRNA target. Here we characterise the use of snoMEN vectors to target the knock-down of micro RNA primary transcripts. We document the specific knock-down of miR21 in HeLa cells using plasmid vectors expressing miR21-targeted snoMEN RNAs and show this induces apoptosis. Knock-down is dependent on the presence of complementary sequences in the snoMEN vector and the induction of apoptosis can be suppressed by over-expression of miR21. Furthermore, we have also developed lentiviral vectors for delivery of snoMEN RNAs and show this increases the efficiency of vector transduction in many human cell lines that are difficult to transfect with plasmid vectors. Transduction of lentiviral vectors expressing snoMEN targeted to pri-miR21 induces apoptosis in human lung adenocarcinoma cells, which express high levels of miR21, but not in human primary cells. We show that snoMEN-mediated suppression of miRNA expression is prevented by siRNA knock-down of Ago2, but not by knock-down of Ago1 or Upf1. snoMEN RNAs colocalise with Ago2 in cell nuclei and nucleoli and can be co-immunoprecipitated from nuclear extracts by antibodies specific for Ago2.


Adenocarcinoma/genetics , Gene Knockdown Techniques/methods , Lung Neoplasms/genetics , MicroRNAs/genetics , RNA Precursors/genetics , RNA, Antisense/genetics , Adenocarcinoma of Lung , Apoptosis , Argonaute Proteins/genetics , Eukaryotic Initiation Factors/genetics , Genetic Vectors/pharmacology , HEK293 Cells , HeLa Cells , Humans , Lentivirus/genetics , Plasmids/genetics , RNA Helicases , Trans-Activators/genetics
12.
Semin Cell Dev Biol ; 44: 107-14, 2015 Aug.
Article En | MEDLINE | ID: mdl-26319183

The embryonic development of the pancreas is orchestrated by a complex and coordinated transcription factor network. Neurogenin3 (Neurog3) initiates the endocrine program by activating the expression of additional transcription factors driving survival, proliferation, maturation and lineage allocation of endocrine precursors. Among the direct targets of Neurog3, Pax4 appears as one of the key regulators of ß-cell specification. Indeed, mice lacking Pax4 die a few days postpartum, as they develop severe hyperglycemia due to the absence of mature pancreatic ß-cells. Pax4 also directly regulates the expression of Arx, a gene that plays a crucial role in α-cell specification. Comparative analysis of Pax4 and Arx mutants, as well as Arx/Pax4 double mutants, showed that islet subtype destiny is mainly directed by cross-repression of the Pax4 and Arx factors. Importantly, the ectopic expression of Pax4 in α-cells was found sufficient to induce their neogenesis and conversion into ß-like cells, not only during development but also in adult rodents. Therefore, differentiated endocrine α-cells can be considered as a putative source for insulin-producing ß-like cells. These findings have clearly widened our understanding regarding pancreatic development, but they also open new research avenues in the context of diabetes research.


Homeodomain Proteins/physiology , Paired Box Transcription Factors/physiology , Animals , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Mice , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , Pancreas/embryology , Pancreas/metabolism , Pancreas/physiology
13.
Hum Gene Ther ; 26(4): 220-31, 2015 Apr.
Article En | MEDLINE | ID: mdl-25758764

Cytokine-induced killer (CIK) cells consist of a heterogeneous population of polyclonal T lymphocytes displaying NK phenotype and HLA-unrestricted cytotoxic activity against a broad range of tumors. We sought to determine whether transduction of CIK cells with T cell receptor (TCR) genes specific for tumor-associated antigens could generate effector cells endowed with a double mechanism of tumor recognition. HLA-A2-restricted TCR-transduced (TD) CIK directed against the melanoma antigens Mart1 and NY-ESO1 were generated by lentiviral transduction and successfully expanded over a 3-4-week period. TD-CIK cells were both CD3(+)/CD56(-) and CD3(+)/CD56(+) (31±8% and 59±9%, respectively), indicating that both major histocompatibility complex (MHC)-restricted T cells and MHC-unrestricted CIK could be targeted by lentiviral transduction. At the end of the culture, the majority of both unmodified and TD-CIK displayed an effector memory phenotype, without considerable expression of replicative senescence and exhaustion markers. Functionally, TD-CIK specifically recognized tumor cells expressing the relevant antigen as well as maintained their MHC-unrestricted tumor activity. The cytotoxic activity of TD-CIK against HLA-A2(+) melanoma cell lines was significantly higher than the untransduced counterparts at a low effector:target ratio (cytotoxic activity of TD-CIK was from 1.9- to 4.3-fold higher than untransduced counterparts). TD-CIK were highly proficient in releasing high amount of IFN-γ upon antigen-specific stimulation and were able to recognize primary melanoma targets. In conclusion, we showed that (1) the reproducibility and simplicity of CIK transduction and expansion might solve the problem of obtaining adequate numbers of potent antitumor effector cells for adoptive immunotherapy; (2) the presence of both terminal effectors as well as of less differentiated progenitors might confer them long survival in vivo; and (3) the addition of an MHC-restricted antigen recognition allows not only targeting tumor surface antigens but also a wider range of cytoplasmic or nuclear antigens, involved in tumor proliferation and survival. TD-CIK cells with a double mechanism of tumor recognition are an attractive and alternative tool for the development of efficient cell therapeutic strategies.


B-Lymphocyte Subsets/immunology , Cytokines/physiology , Melanoma/therapy , Natural Killer T-Cells/immunology , Receptors, Antigen, T-Cell/biosynthesis , Antigens, Neoplasm/immunology , Cell Line, Tumor , Genetic Engineering , Humans , Immunotherapy, Adoptive , Leukocytes, Mononuclear/immunology , Melanoma/immunology , Receptors, Antigen, T-Cell/genetics
14.
Stem Cells ; 32(3): 754-69, 2014 Mar.
Article En | MEDLINE | ID: mdl-24123662

Signals downstream of Akt can either favor or oppose stem cell (SC) maintenance, but how this dual role can be achieved is still undefined. Using human limbal keratinocyte stem cells (LKSCs), a SC type used in transplantation therapies for corneal regeneration, we show that Akt signaling is prominent in SC populations both in vivo and in vitro, and that Akt1 promotes while Akt2 opposes SC self-renewal. Noteworthy, loss of Akt2 signaling enhances LKSC maintenance ex vivo, whereas Akt1 depletion anticipates SC exhaustion. Mechanistically, the antagonistic functions of Akt1 and Akt2 in SC control are mainly dictated by their differential subcellular distribution, being nuclear Akt2 selectively implicated in FOXO inhibition. Akt2 downregulation favors LKSC maintenance as a result of a gain of FOXO functions, which attenuates the mechanistic target of rapamycin complex one signaling via tuberous sclerosis one gene induction, and promotes growth factor signaling through Akt1. Consistently, Akt2 deficiency also enhances limbal SCs in vivo. Thus, our findings reveal distinct roles for nuclear versus cytosolic Akt signaling in normal epithelial SC control and suggest that the selective Akt2 inhibition may provide novel pharmacological strategies for human LKSC expansion in therapeutic settings and mechanistic research.


Cell Nucleus/enzymology , Forkhead Transcription Factors/metabolism , Keratinocytes/cytology , Multiprotein Complexes/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Stem Cells/cytology , TOR Serine-Threonine Kinases/metabolism , 3T3 Cells , Adult , Animals , Cell Proliferation , Clone Cells , Enzyme Activation , Forkhead Box Protein O1 , Forkhead Box Protein O3 , Humans , Isoenzymes/metabolism , Limbus Corneae/cytology , Mechanistic Target of Rapamycin Complex 1 , Mice , Phenotype , Phosphorylation , Proto-Oncogene Proteins c-akt/deficiency , Repressor Proteins/metabolism , Signal Transduction , Stem Cells/enzymology , Transcription, Genetic
15.
Curr Top Dev Biol ; 106: 217-38, 2013.
Article En | MEDLINE | ID: mdl-24290351

Type 1 diabetes is a metabolic disease resulting in the selective loss of pancreatic insulin-producing ß-cells and affecting millions of people worldwide. The side effects of diabetes are varied and include cardiovascular, neuropathologic, and kidney diseases. Despite the most recent advances in diabetes care, patients suffering from type 1 diabetes still display a shortened life expectancy compared to their healthy counterparts. In an effort to improve ß-cell-replacement therapies, numerous approaches are currently being pursued, most of these aiming at finding ways to differentiate stem/progenitor cells into ß-like cells by mimicking embryonic development. Unfortunately, these efforts have hitherto not allowed the generation of fully functional ß-cells. This chapter summarizes recent findings, allowing a better insight into the molecular mechanisms underlying the genesis of ß-cells during the course of pancreatic morphogenesis. Furthermore, a focus is made on new research avenues concerning the conversion of pre-existing pancreatic cells into ß-like cells, such approaches holding great promise for the development of type 1 diabetes therapies.


Embryonic Stem Cells/physiology , Insulin-Secreting Cells/physiology , Pancreas/embryology , Regeneration , Animals , Cell Dedifferentiation/physiology , Cell Differentiation/physiology , Diabetes Mellitus, Type 1/surgery , Embryonic Stem Cells/cytology , Embryonic Stem Cells/transplantation , Humans , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/transplantation , Pancreas/cytology , Pancreas/growth & development , Regenerative Medicine/methods
16.
PLoS Genet ; 9(10): e1003934, 2013 Oct.
Article En | MEDLINE | ID: mdl-24204325

Recently, it was demonstrated that pancreatic new-born glucagon-producing cells can regenerate and convert into insulin-producing ß-like cells through the ectopic expression of a single gene, Pax4. Here, combining conditional loss-of-function and lineage tracing approaches, we show that the selective inhibition of the Arx gene in α-cells is sufficient to promote the conversion of adult α-cells into ß-like cells at any age. Interestingly, this conversion induces the continuous mobilization of duct-lining precursor cells to adopt an endocrine cell fate, the glucagon(+) cells thereby generated being subsequently converted into ß-like cells upon Arx inhibition. Of interest, through the generation and analysis of Arx and Pax4 conditional double-mutants, we provide evidence that Pax4 is dispensable for these regeneration processes, indicating that Arx represents the main trigger of α-cell-mediated ß-like cell neogenesis. Importantly, the loss of Arx in α-cells is sufficient to regenerate a functional ß-cell mass and thereby reverse diabetes following toxin-induced ß-cell depletion. Our data therefore suggest that strategies aiming at inhibiting the expression of Arx, or its molecular targets/co-factors, may pave new avenues for the treatment of diabetes.


Cell Differentiation , Diabetes Mellitus, Type 1/genetics , Homeodomain Proteins/genetics , Insulin-Secreting Cells/metabolism , Transcription Factors/genetics , Animals , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/therapy , Disease Models, Animal , Gene Expression Regulation , Glucagon/genetics , Glucagon/metabolism , Glucagon-Secreting Cells/metabolism , Glucagon-Secreting Cells/pathology , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/biosynthesis , Humans , Insulin-Secreting Cells/cytology , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Mice, Transgenic , Paired Box Transcription Factors/genetics , Transcription Factors/antagonists & inhibitors , Transcription Factors/biosynthesis
17.
Med Sci (Paris) ; 29(8-9): 749-55, 2013.
Article Fr | MEDLINE | ID: mdl-24005630

Type 1 diabetes (T1DM) is a common metabolic disorder affecting an ever-increasing number of patients worldwide. T1DM is caused by the selective destruction of pancreatic insulin-producing ß-cells by the immune system. Such loss results in chronic hyperglycemia and could induce a number of cardio-vascular complications. Despite the classical insulin-based therapy, compared to healthy people, patients with T1DM display a shortened life expectancy due to the treatment's inability to strictly regulate glycemic levels. An alternative therapy involves pancreatic islet transplantation but the shortage of donors and the required immuno-suppressive treatments limit the widespread use of this approach. Therefore, the search of new approaches to generate functional ß-cells is of growing interest. In this review, we describe several novel strategies aiming at the conversion of diverse pancreatic cells into ß-cells, such as acinar, ductal, and endocrine cells. Clearly, such promising results could open new research avenues in the context of type 1 diabetes research.


Cell Differentiation , Diabetes Mellitus, Type 1/therapy , Insulin-Secreting Cells/cytology , Pancreas/cytology , Acinar Cells/cytology , Diabetes Mellitus, Type 1/surgery , Humans , Insulin-Secreting Cells/physiology , Islets of Langerhans Transplantation , Pancreatic Ducts/cytology , Regeneration , Tissue Donors/supply & distribution
19.
Dev Cell ; 26(1): 86-100, 2013 Jul 15.
Article En | MEDLINE | ID: mdl-23810513

It was recently demonstrated that embryonic glucagon-producing cells in the pancreas can regenerate and convert into insulin-producing ß-like cells through the constitutive/ectopic expression of the Pax4 gene. However, whether α cells in adult mice display the same plasticity is unknown. Similarly, the mechanisms underlying such reprogramming remain unclear. We now demonstrate that the misexpression of Pax4 in glucagon(+) cells age-independently induces their conversion into ß-like cells and their glucagon shortage-mediated replacement, resulting in islet hypertrophy and in an unexpected islet neogenesis. Combining several lineage-tracing approaches, we show that, upon Pax4-mediated α-to-ß-like cell conversion, pancreatic duct-lining precursor cells are continuously mobilized, re-express the developmental gene Ngn3, and successively adopt a glucagon(+) and a ß-like cell identity through a mechanism involving the reawakening of the epithelial-to-mesenchymal transition. Importantly, these processes can repeatedly regenerate the whole ß cell mass and thereby reverse several rounds of toxin-induced diabetes, providing perspectives to design therapeutic regenerative strategies.


Cellular Reprogramming , Diabetes Mellitus, Experimental/metabolism , Insulin-Secreting Cells/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Blood Glucose/analysis , Cell Differentiation , Cell Lineage , Cell Movement , Diabetes Mellitus, Experimental/genetics , Epithelial-Mesenchymal Transition , Gene Expression Regulation , Glucagon-Secreting Cells/metabolism , Glucagon-Secreting Cells/pathology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Hypertrophy/metabolism , Hypertrophy/pathology , Insulin-Secreting Cells/pathology , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , Pancreatic Ducts/drug effects , Pancreatic Ducts/metabolism , Pancreatic Ducts/pathology , Streptozocin
20.
PLoS One ; 8(4): e62305, 2013.
Article En | MEDLINE | ID: mdl-23638031

The study of the function of many human proteins is often hampered by technical limitations, such as cytotoxicity and phenotypes that result from overexpression of the protein of interest together with the endogenous version. Here we present the snoMEN (snoRNA Modulator of gene ExpressioN) vector technology for generating stable cell lines where expression of the endogenous protein can be reduced and replaced by an exogenous protein, such as a fluorescent protein (FP)-tagged version. SnoMEN are snoRNAs engineered to contain complementary sequences that can promote knock-down of targeted RNAs. We have established and characterised two such partial protein replacement human cell lines (snoMEN-PR). Quantitative mass spectrometry was used to analyse the specificity of knock-down and replacement at the protein level and also showed an increased pull-down efficiency of protein complexes containing exogenous, tagged proteins in the protein replacement cell lines, as compared with conventional co-expression strategies. The snoMEN approach facilitates the study of mammalian proteins, particularly those that have so far been difficult to investigate by exogenous expression and has wide applications in basic and applied gene-expression research.


Genetic Engineering/methods , Genetic Vectors/genetics , Proteins/metabolism , RNA, Small Nucleolar/genetics , Cell Line , Gene Expression , Humans , Proteins/genetics , RNA Interference , RNA, Small Interfering/genetics
...