Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Nat Commun ; 15(1): 3318, 2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38632302

ABSTRACT

Pancreatic islets of Langerhans play a pivotal role in regulating blood glucose homeostasis, but critical information regarding their mass, distribution and composition is lacking within a whole organ context. Here, we apply a 3D imaging pipeline to generate a complete account of the insulin-producing islets throughout the human pancreas at a microscopic resolution and within a maintained spatial 3D context. These data show that human islets are far more heterogenous than previously accounted for with regards to their size distribution and cellular make up. By deep tissue 3D imaging, this in-depth study demonstrates that 50% of the human insulin-expressing islets are virtually devoid of glucagon-producing α-cells, an observation with significant implications for both experimental and clinical research.


Subject(s)
Glucagon-Secreting Cells , Islets of Langerhans , Humans , Pancreas/metabolism , Islets of Langerhans/metabolism , Insulin/metabolism , Glucagon-Secreting Cells/metabolism , Blood Glucose/metabolism , Insulin Secretion
2.
Front Neurosci ; 18: 1328815, 2024.
Article in English | MEDLINE | ID: mdl-38601090

ABSTRACT

Introduction: Optical Projection Tomography (OPT) and light sheet fluorescence microscopy (LSFM) are high resolution optical imaging techniques, ideally suited for ex vivo 3D whole mouse brain imaging. Although they exhibit high specificity for their targets, the anatomical detail provided by tissue autofluorescence remains limited. Methods: T1-weighted images were acquired from 19 BABB or DBE cleared brains to create an MR template using serial longitudinal registration. Afterwards, fluorescent OPT and LSFM images were coregistered/normalized to the MR template to create fusion images. Results: Volumetric calculations revealed a significant difference between BABB and DBE cleared brains, leading to develop two optimized templates, with associated tissue priors and brain atlas, for BABB (OCUM) and DBE (iOCUM). By creating fusion images, we identified virus infected brain regions, mapped dopamine transporter and translocator protein expression, and traced innervation from the eye along the optic tract to the thalamus and superior colliculus using cholera toxin B. Fusion images allowed for precise anatomical identification of fluorescent signal in the detailed anatomical context provided by MR. Discussion: The possibility to anatomically map fluorescent signals on magnetic resonance (MR) images, widely used in clinical and preclinical neuroscience, would greatly benefit applications of optical imaging of mouse brain. These specific MR templates for cleared brains enable a broad range of neuroscientific applications integrating 3D optical brain imaging.

3.
Nat Commun ; 14(1): 2007, 2023 04 10.
Article in English | MEDLINE | ID: mdl-37037810

ABSTRACT

Viral tropism within the brain and the role(s) of vertebrate immune response to neurotropic flaviviruses infection is largely understudied. We combine multimodal imaging (cm-nm scale) with single nuclei RNA-sequencing to study Langat virus in wildtype and interferon alpha/beta receptor knockout (Ifnar-/-) mice to visualize viral pathogenesis and define molecular mechanisms. Whole brain viral infection is imaged by Optical Projection Tomography coregistered to ex vivo MRI. Infection is limited to grey matter of sensory systems in wildtype mice, but extends into white matter, meninges and choroid plexus in Ifnar-/- mice. Cells in wildtype display strong type I and II IFN responses, likely due to Ifnb expressing astrocytes, infiltration of macrophages and Ifng-expressing CD8+ NK cells, whereas in Ifnar-/-, the absence of this response contributes to a shift in cellular tropism towards non-activated resident microglia. Multimodal imaging-transcriptomics exemplifies a powerful way to characterize mechanisms of viral pathogenesis and tropism.


Subject(s)
Encephalitis Viruses, Tick-Borne , Interferon Type I , Ticks , Mice , Animals , Interferon Type I/metabolism , Neurons/metabolism , Mice, Knockout , Brain/diagnostic imaging , Brain/metabolism , Encephalitis Viruses, Tick-Borne/genetics , Encephalitis Viruses, Tick-Borne/metabolism , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , Tropism , Ticks/metabolism , Mice, Inbred C57BL
4.
Methods Mol Biol ; 2592: 1-19, 2023.
Article in English | MEDLINE | ID: mdl-36507982

ABSTRACT

The rodent pancreas is the prevalent model system for preclinical diabetes research. However, due to the compound endocrine-exocrine organization of the gland, with the endocrine islets of Langerhans scattered by the thousands throughout the much greater exocrine parenchyma, stereological assessments of endocrine cell mass, commonly insulin-producing ß-cells, are exceedingly challenging. In recent years, optical mesoscopic imaging techniques such as optical projection tomography (OPT) and light sheet fluorescence microscopy (LSFM) have seen dramatic developments, enabling 3D visualization of fluorescently labeled cells in mm- to cm-sized tissues with µm resolution. Here we present a protocol for 3D visualization and "absolute" quantitative assessments of, for example, islet mass throughout the volume of rodent pancreata with maintained spatial context.


Subject(s)
Islets of Langerhans , Tomography, Optical , Animals , Rodentia , Tomography, Optical/methods , Pancreas/diagnostic imaging , Microscopy, Fluorescence , Molecular Imaging , Islets of Langerhans/diagnostic imaging , Imaging, Three-Dimensional/methods
5.
Sci Data ; 9(1): 558, 2022 09 10.
Article in English | MEDLINE | ID: mdl-36088402

ABSTRACT

Mouse models for streptozotocin (STZ) induced diabetes probably represent the most widely used systems for preclinical diabetes research, owing to the compound's toxic effect on pancreatic ß-cells. However, a comprehensive view of pancreatic ß-cell mass distribution subject to STZ administration is lacking. Previous assessments have largely relied on the extrapolation of stereological sections, which provide limited 3D-spatial and quantitative information. This data descriptor presents multiple ex vivo tomographic optical image datasets of the full ß-cell mass distribution in mice subject to single high and multiple low doses of STZ administration, and in glycaemia recovered mice. The data further include information about structural features, such as individual islet ß-cell volumes, spatial coordinates, and shape as well as signal intensities for both insulin and GLUT2. Together, they provide the most comprehensive anatomical record of the effects of STZ administration on the islet of Langerhans in mice. As such, this data descriptor may serve as reference material to facilitate the planning, use and (re)interpretation of this widely used disease model.


Subject(s)
Diabetes Mellitus, Experimental , Islets of Langerhans , Animals , Blood Glucose/analysis , Insulin/analysis , Mice , Streptozocin/analysis
6.
Mol Pharm ; 18(12): 4428-4436, 2021 12 06.
Article in English | MEDLINE | ID: mdl-34649437

ABSTRACT

Type 1 diabetes develops in childhood and adolescence, with peak incidence in the early teenage years. There is an urgent need for an accurate method to detect insulin-producing ß-cells in patients that is not affected by alterations in ß-cell function. As part of our research program to design specific probes to measure ß-cell mass, we recently developed a novel insulin-binding peptide probe (IBPP) for the detection of ß-cells in vivo. Here, we applied our innovative method to show specific labeling of this IBPP to human and mouse fixed ß-cells in pancreatic islets. Importantly, we showed staining of human and mouse islets in culture without any negative functional or cell viability impact. Moreover, the IBPP-stained mouse islets after tail vein injection in vivo, albeit with batch differences in staining efficiency. In conclusion, we provide evidence showing that the IBPP can be used for future accurate detection of ß-cell mass in a variety of preclinical models of diabetes.


Subject(s)
Diabetes Mellitus, Type 1/diagnostic imaging , Insulin-Secreting Cells/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Animals , Cells, Cultured , Humans , Insulin/analysis , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Staining and Labeling
7.
Commun Biol ; 4(1): 1063, 2021 09 10.
Article in English | MEDLINE | ID: mdl-34508173

ABSTRACT

The possibility to quantitatively study specific molecular/cellular features of complete human organs with preserved spatial 3D context would have widespread implications for pre-clinical and clinical medicine. Whereas optical 3D imaging approaches have experienced a formidable revolution, they have remained limited due to current incapacities in obtaining specific labelling within large tissue volumes. We present a simple approach enabling reconstruction of antibody labeled cells within entire human organs with preserved organ context. We demonstrate the utility of the approach by providing volumetric data and 3D distribution of hundreds of thousands of islets of Langerhans within the human pancreas. By assessments of pancreata from non-diabetic and type 2 diabetic individuals, we display previously unrecognized features of the human islet mass distribution and pathology. As such, this method may contribute not only in unraveling new information of the pancreatic anatomy/pathophysiology, but it may be translated to essentially any antibody marker or organ system.


Subject(s)
Imaging, Three-Dimensional , Islets of Langerhans/cytology , Aged , Humans , Male
8.
eNeuro ; 8(5)2021.
Article in English | MEDLINE | ID: mdl-34417283

ABSTRACT

Opsin 3 (Opn3) is highly expressed in the adult brain, however, information for spatial and temporal expression patterns during embryogenesis is significantly lacking. Here, an Opn3-eGFP reporter mouse line was used to monitor cell body expression and axonal projections during embryonic and early postnatal to adult stages. By applying 2D and 3D fluorescence imaging techniques, we have identified the onset of Opn3 expression, which predominantly occurred during embryonic stages, in various structures during brain/head development. In addition, this study defines over twenty Opn3-eGFP-positive neural structures never reported before. Opn3-eGFP was first observed at E9.5 in neural regions, including the ganglia that will ultimately form the trigeminal, facial and vestibulocochlear cranial nerves (CNs). As development proceeds, expanded Opn3-eGFP expression coincided with the formation and maturation of critical components of the central and peripheral nervous systems (CNS, PNS), including various motor-sensory tracts, such as the dorsal column-medial lemniscus (DCML) sensory tract, and olfactory, acoustic, and optic tracts. The widespread, yet distinct, detection of Opn3-eGFP already at early embryonic stages suggests that Opn3 might play important functional roles in the developing brain and spinal cord to regulate multiple motor and sensory circuitry systems, including proprioception, nociception, ocular movement, and olfaction, as well as memory, mood, and emotion. This study presents a crucial blueprint from which to investigate autonomic and cognitive opsin-dependent neural development and resultant behaviors under physiological and pathophysiological conditions.


Subject(s)
Opsins , Rod Opsins , Animals , Embryo, Mammalian , Embryonic Development , Mice , Spinal Cord
9.
Mol Metab ; 53: 101264, 2021 11.
Article in English | MEDLINE | ID: mdl-34091063

ABSTRACT

OBJECTIVE: Early postnatal life is a critical period for the establishment of the functional ß-cell mass that will sustain whole-body glucose homeostasis during the lifetime. ß cells are formed from progenitors during embryonic development but undergo significant expansion in quantity and attain functional maturity after birth. The signals and pathways involved in these processes are not fully elucidated. Cyclic adenosine monophosphate (cAMP) is an intracellular signaling molecule that is known to regulate insulin secretion, gene expression, proliferation, and survival of adult ß cells. The heterotrimeric G protein Gs stimulates the cAMP-dependent pathway by activating adenylyl cyclase. In this study, we sought to explore the role of Gs-dependent signaling in postnatal ß-cell development. METHODS: To study Gs-dependent signaling, we generated conditional knockout mice in which the α subunit of the Gs protein (Gsα) was ablated from ß-cells using the Cre deleter line Ins1Cre. Mice were characterized in terms of glucose homeostasis, including in vivo glucose tolerance, glucose-induced insulin secretion, and insulin sensitivity. ß-cell mass was studied using histomorphometric analysis and optical projection tomography. ß-cell proliferation was studied by ki67 and phospho-histone H3 immunostatining, and apoptosis was assessed by TUNEL assay. Gene expression was determined in isolated islets and sorted ß cells by qPCR. Intracellular cAMP was studied in isolated islets using HTRF-based technology. The activation status of the cAMP and insulin-signaling pathways was determined by immunoblot analysis of the relevant components of these pathways in isolated islets. In vitro proliferation of dissociated islet cells was assessed by BrdU incorporation. RESULTS: Elimination of Gsα in ß cells led to reduced ß-cell mass, deficient insulin secretion, and severe glucose intolerance. These defects were evident by weaning and were associated with decreased proliferation and inadequate expression of key ß-cell identity and maturation genes in postnatal ß-cells. Additionally, loss of Gsα caused a broad multilevel disruption of the insulin transduction pathway that resulted in the specific abrogation of the islet proliferative response to insulin. CONCLUSION: We conclude that Gsα is required for ß-cell growth and maturation in the early postnatal stage and propose that this is partly mediated via its crosstalk with insulin signaling. Our findings disclose a tight connection between these two pathways in postnatal ß cells, which may have implications for using cAMP-raising agents to promote ß-cell regeneration and maturation in diabetes.


Subject(s)
GTP-Binding Protein alpha Subunits, Gs/metabolism , Insulin-Secreting Cells/metabolism , Animals , GTP-Binding Protein alpha Subunits, Gs/deficiency , Mice, Knockout , Mice, Transgenic , Signal Transduction
10.
Front Endocrinol (Lausanne) ; 12: 633063, 2021.
Article in English | MEDLINE | ID: mdl-33746904

ABSTRACT

The exocrine-endocrine multipart organization of the pancreas makes it an exceedingly challenging organ to analyze, quantitatively and spatially. Both in rodents and humans, estimates of the pancreatic cellular composition, including beta-cell mass, has been largely relying on the extrapolation of 2D stereological data originating from limited sample volumes. Alternatively, they have been obtained by low resolution non-invasive imaging techniques providing little detail regarding the anatomical organization of the pancreas and its cellular and/or molecular make up. In this mini-review, the state of the art and the future potential of currently existing and emerging high-resolution optical imaging techniques working in the mm-cm range with µm resolution, here referred to as mesoscopic imaging approaches, will be discussed regarding their contribution toward a better understanding of pancreatic anatomy both in normal conditions and in the diabetic setting. In particular, optical projection tomography (OPT) and light sheet fluorescence microscopy (LSFM) imaging of the pancreas and their associated tissue processing and computational analysis protocols will be discussed in the light of their current capabilities and future potential to obtain more detailed 3D-spatial, quantitative, and molecular information of the pancreas.


Subject(s)
Microscopy, Fluorescence/methods , Optical Imaging/methods , Pancreas/diagnostic imaging , Tomography, Optical/methods , Humans , Imaging, Three-Dimensional/methods
11.
J Pathol ; 254(1): 31-45, 2021 05.
Article in English | MEDLINE | ID: mdl-33527355

ABSTRACT

Maturity-onset diabetes of the young type 5 (MODY5) is due to heterozygous mutations or deletion of HNF1B. No mouse models are currently available to recapitulate the human MODY5 disease. Here, we investigate the pancreatic phenotype of a unique MODY5 mouse model generated by heterozygous insertion of a human HNF1B splicing mutation at the intron-2 splice donor site in the mouse genome. This Hnf1bsp2/+ model generated with targeted mutation of Hnf1b mimicking the c.544+1G>T (T) mutation identified in humans, results in alternative transcripts and a 38% decrease of native Hnf1b transcript levels. As a clinical feature of MODY5 patients, the hypomorphic mouse model Hnf1bsp2/+ displays glucose intolerance. Whereas Hnf1bsp2/+ isolated islets showed no altered insulin secretion, we found a 65% decrease in pancreatic insulin content associated with a 30% decrease in total large islet volume and a 20% decrease in total ß-cell volume. These defects were associated with a 30% decrease in expression of the pro-endocrine gene Neurog3 that we previously identified as a direct target of Hnf1b, showing a developmental etiology. As another clinical feature of MODY5 patients, the Hnf1bsp2/+ pancreases display exocrine dysfunction with hypoplasia. We observed chronic pancreatitis with loss of acinar cells, acinar-to-ductal metaplasia, and lipomatosis, with upregulation of signaling pathways and impaired acinar cell regeneration. This was associated with ductal cell deficiency characterized by shortened primary cilia. Importantly, the Hnf1bsp2/+ mouse model reproduces the pancreatic features of the human MODY5/HNF1B disease, providing a unique in vivo tool for molecular studies of the endocrine and exocrine defects and to advance basic and translational research. © 2021 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.


Subject(s)
Central Nervous System Diseases/genetics , Central Nervous System Diseases/physiopathology , Dental Enamel/abnormalities , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/physiopathology , Disease Models, Animal , Hepatocyte Nuclear Factor 1-beta/genetics , Kidney Diseases, Cystic/genetics , Kidney Diseases, Cystic/physiopathology , Pancreas/physiopathology , Animals , Central Nervous System Diseases/pathology , Dental Enamel/pathology , Dental Enamel/physiopathology , Diabetes Mellitus, Type 2/pathology , Humans , Kidney Diseases, Cystic/pathology , Mice , Mice, Transgenic , Mutation , Pancreas/pathology , Phenotype
12.
Trends Endocrinol Metab ; 31(12): 905-917, 2020 12.
Article in English | MEDLINE | ID: mdl-33160815

ABSTRACT

A common feature in the pathophysiology of different types of diabetes is the reduction of ß cell mass and/or impairment of ß cell function. Diagnosis and treatment of type 1 and type 2 diabetes is currently hampered by a lack of reliable techniques to restore ß cell survival, to improve insulin secretion, and to quantify ß cell mass in patients. Current new approaches may allow us to precisely and specifically visualize ß cells in vivo and provide viable therapeutic strategies to preserve, recover, and regenerate ß cells. In this review, we discuss recent protective approaches for ß cells and the advantages and limitations of current imaging probes in the field.


Subject(s)
Diabetes Mellitus, Type 1/physiopathology , Diabetes Mellitus, Type 2/physiopathology , Animals , Humans , Insulin-Secreting Cells/physiology
13.
Sci Rep ; 10(1): 18246, 2020 10 26.
Article in English | MEDLINE | ID: mdl-33106532

ABSTRACT

The possibility to assess pancreatic anatomy with microscopic resolution in three dimensions (3D) would significantly add to pathological analyses of disease processes. Pancreatic ductal adenocarcinoma (PDAC) has a bleak prognosis with over 90% of the patients dying within 5 years after diagnosis. Cure can be achieved by surgical resection, but the efficiency remains drearily low. Here we demonstrate a method that without prior immunohistochemical labelling provides insight into the 3D microenvironment and spread of PDAC and premalignant cysts in intact surgical biopsies. The method is based solely on the autofluorescent properties of the investigated tissues using optical projection tomography and/or light-sheet fluorescence microscopy. It does not interfere with subsequent histopathological analysis and may facilitate identification of tumor-free resection margins within hours. We further demonstrate how the developed approach can be used to assess individual volumes and numbers of the islets of Langerhans in unprecedently large biopsies of human pancreatic tissue, thus providing a new means by which remaining islet mass may be assessed in settings of diabetes. Generally, the method may provide a fast approach to provide new anatomical insight into pancreatic pathophysiology.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Islets of Langerhans/pathology , Pancreatic Neoplasms/pathology , Carcinoma, Pancreatic Ductal/diagnostic imaging , Humans , Imaging, Three-Dimensional/methods , Islets of Langerhans/diagnostic imaging , Optical Imaging/methods , Pancreatic Neoplasms/diagnostic imaging , Tomography, Optical/methods , Tumor Microenvironment
14.
Commun Biol ; 3(1): 541, 2020 09 30.
Article in English | MEDLINE | ID: mdl-32999405

ABSTRACT

Mouse models of Streptozotocin (STZ) induced diabetes represent the most widely used preclinical diabetes research systems. We applied state of the art optical imaging schemes, spanning from single islet resolution to the whole organ, providing a first longitudinal, 3D-spatial and quantitative account of ß-cell mass (BCM) dynamics and islet longevity in STZ-treated mice. We demonstrate that STZ-induced ß-cell destruction predominantly affects large islets in the pancreatic core. Further, we show that hyperglycemic STZ-treated mice still harbor a large pool of remaining ß-cells but display pancreas-wide downregulation of glucose transporter type 2 (GLUT2). Islet gene expression studies confirmed this downregulation and revealed impaired ß-cell maturity. Reversing hyperglycemia by islet transplantation partially restored the expression of markers for islet function, but not BCM. Jointly our results indicate that STZ-induced hyperglycemia results from ß-cell dysfunction rather than ß-cell ablation and that hyperglycemia in itself sustains a negative feedback loop restraining islet function recovery.


Subject(s)
Diabetes Mellitus, Experimental/pathology , Insulin-Secreting Cells/pathology , Islets of Langerhans/pathology , Animals , Biomarkers/metabolism , Diabetes Mellitus, Experimental/metabolism , Down-Regulation , Glucose Transporter Type 2/metabolism , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/ultrastructure , Islets of Langerhans/metabolism , Male , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Microscopy, Electron, Transmission , Microscopy, Fluorescence
16.
Diabetologia ; 61(4): 896-905, 2018 04.
Article in English | MEDLINE | ID: mdl-29209740

ABSTRACT

AIMS/HYPOTHESIS: Genetic studies show coupling of genes affecting beta cell function to type 1 diabetes, but hitherto no studies on whether beta cell dysfunction could precede insulitis and clinical onset of type 1 diabetes are available. METHODS: We used 40-day-old BioBreeding (BB) DRLyp/Lyp rats (a model of spontaneous autoimmune type 1 diabetes) and diabetes-resistant DRLyp/+ and DR+/+ littermates (controls) to investigate beta cell function in vivo, and insulin and glucagon secretion in vitro. Beta cell mass was assessed by optical projection tomography (OPT) and morphometry. Additionally, measurements of intra-islet blood flow were performed using microsphere injections. We also assessed immune cell infiltration, cytokine expression in islets (by immunohistochemistry and qPCR), as well as islet Glut2 expression and ATP/ADP ratio to determine effects on glucose uptake and metabolism in beta cells. RESULTS: DRLyp/Lyp rats were normoglycaemic and without traces of immune cell infiltrates. However, IVGTTs revealed a significant decrease in the acute insulin response to glucose compared with control rats (1685.3 ± 121.3 vs 633.3 ± 148.7; p < 0.0001). In agreement, insulin secretion was severely perturbed in isolated islets, and both first- and second-phase insulin release were lowered compared with control rats, while glucagon secretion was similar in both groups. Interestingly, after 5-7 days of culture of islets from DRLyp/Lyp rats in normal media, glucose-stimulated insulin secretion (GSIS) was improved; although, a significant decrease in GSIS was still evident compared with islets from control rats at this time (7393.9 ± 1593.7 vs 4416.8 ± 1230.5 pg islet-1 h-1; p < 0.0001). Compared with controls, OPT of whole pancreas from DRLyp/Lyp rats revealed significant reductions in medium (4.1 × 109 ± 9.5 × 107 vs 3.8 × 109 ± 5.8 × 107 µm3; p = 0.044) and small sized islets (1.6 × 109 ± 5.1 × 107 vs 1.4 × 109 ± 4.5 × 107 µm3; p = 0.035). Finally, we found lower intra-islet blood perfusion in vivo (113.1 ± 16.8 vs 76.9 ± 11.8 µl min-1 [g pancreas]-1; p = 0.023) and alterations in the beta cell ATP/ADP ratio in DRLyp/Lyp rats vs control rats. CONCLUSIONS/INTERPRETATION: The present study identifies a deterioration of beta cell function and mass, and intra-islet blood flow that precedes insulitis and diabetes development in animals prone to autoimmune type 1 diabetes. These underlying changes in islet function may be previously unrecognised factors of importance in type 1 diabetes development.


Subject(s)
Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/metabolism , Disease Models, Animal , Insulin-Secreting Cells/cytology , Insulin/metabolism , Adenosine Diphosphate/chemistry , Adenosine Triphosphate/chemistry , Animals , Blood Glucose/metabolism , Female , Genotype , Glucose/metabolism , Islets of Langerhans/metabolism , Langerhans Cells/metabolism , Male , Pancreas/metabolism , Perfusion , Rats , Rats, Inbred BB , Rats, Wistar
17.
Sci Rep ; 7(1): 12440, 2017 09 29.
Article in English | MEDLINE | ID: mdl-28963457

ABSTRACT

Functional beta cell mass is an essential biomarker for the diagnosis and staging of diabetes. It has however proven technically challenging to study this parameter during diabetes progression. Here we have detailed the kinetics of the rapid decline in functional beta cell mass in the RIP-DTR mouse, a model of hyperglycemia resulting from diphtheria toxin induced beta cell ablation. A novel combination of imaging modalities was employed to study the pattern of beta cell destruction. Optical projection tomography of the pancreas and longitudinal in vivo confocal microscopy of islets transplanted into the anterior chamber of the eye allowed to investigate kinetics and tomographic location of beta cell mass decay in individual islets as well as at the entire islet population level. The correlation between beta cell mass and function was determined by complementary in vivo and ex vivo characterizations, demonstrating that beta cell function and glucose tolerance were impaired within the first two days following treatment when more than 50% of beta cell mass was remaining. Our results illustrate the importance of acquiring quantitative functional and morphological parameters to assess the functional status of the endocrine pancreas.


Subject(s)
Diabetes Mellitus, Experimental/pathology , Heparin-binding EGF-like Growth Factor/genetics , Hyperglycemia/pathology , Insulin/deficiency , Islets of Langerhans/ultrastructure , Recombinant Fusion Proteins/genetics , Animals , Anterior Chamber/surgery , Blood Glucose/metabolism , Cell Count , Cell Death , Choristoma , Diabetes Mellitus, Experimental/diagnostic imaging , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Gene Expression , Glucose Tolerance Test , Heparin-binding EGF-like Growth Factor/metabolism , Hyperglycemia/diagnostic imaging , Hyperglycemia/genetics , Hyperglycemia/metabolism , Insulin/genetics , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Islets of Langerhans Transplantation/methods , Male , Mice , Mice, Transgenic , Microscopy, Confocal , Promoter Regions, Genetic , Rats , Recombinant Fusion Proteins/metabolism , Tomography, Optical
18.
Sci Rep ; 7(1): 6646, 2017 07 27.
Article in English | MEDLINE | ID: mdl-28751653

ABSTRACT

Despite the dramatic increase in the prevalence of diabetes, techniques for in situ studies of the underlying pancreatic biochemistry are lacking. Such methods would facilitate obtaining mechanistic understanding of diabetes pathophysiology and aid in prognostic and/or diagnostic assessments. In this report we demonstrate how a multivariate imaging approach (orthogonal projections to latent structures - discriminant analysis) can be applied to generate full vibrational microspectroscopic profiles of pancreatic tissues. These profiles enable extraction of known and previously unrecorded biochemical alterations in models of diabetes, and allow for classification of the investigated tissue with regards to tissue type, strain and stage of disease progression. Most significantly, the approach provided evidence for dramatic alterations of the pancreatic biochemistry at the initial onset of immune-infiltration in the Non Obese Diabetic model for type 1 diabetes. Further, it enabled detection of a previously undocumented accumulation of collagen fibrils in the leptin deficient ob/ob mouse islets. By generating high quality spectral profiles through the tissue capsule of hydrated human pancreata and by in vivo Raman imaging of pancreatic islets transplanted to the anterior chamber of the eye, we provide critical feasibility studies for the translation of this technique to diagnostic assessments of pancreatic biochemistry in vivo.


Subject(s)
Diabetes Mellitus, Type 1/diagnosis , Pancreas/metabolism , Spectrum Analysis/methods , Animals , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/physiopathology , Disease Models, Animal , Disease Progression , Female , Humans , Leptin/genetics , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Pancreas/physiopathology
19.
Sci Data ; 4: 170031, 2017 03 14.
Article in English | MEDLINE | ID: mdl-28291266

ABSTRACT

A detailed understanding of pancreatic ß-cell mass distribution is a key element to fully appreciate the pathophysiology of models of diabetes and metabolic stress. Commonly, such assessments have been performed by stereological approaches that rely on the extrapolation of two-dimensional data and provide very limited topological information. We present ex vivo optical tomographic data sets of the full ß-cell mass distribution in cohorts of obese ob/ob mice and their lean controls, together with information about individual islet ß-cell volumes, their three-dimensional coordinates and shape throughout the volume of the pancreas between 4 and 52 weeks of age. These data sets offer the currently most comprehensive public record of the ß-cell mass distribution in the mouse. As such, they may serve as a quantitative and topological reference for the planning of a variety of in vivo or ex vivo experiments including computational modelling and statistical analyses. By shedding light on intra- and inter-lobular variations in ß-cell mass distribution, they further provide a powerful tool for the planning of stereological sampling assessments.


Subject(s)
Islets of Langerhans/diagnostic imaging , Animals , Mice , Mice, Obese , Species Specificity , Tomography, Optical
20.
Sci Rep ; 6: 34885, 2016 10 07.
Article in English | MEDLINE | ID: mdl-27713548

ABSTRACT

The leptin deficient ob/ob mouse is a widely used model for studies on initial aspects of metabolic disturbances leading to type 2 diabetes, including insulin resistance and obesity. Although it is generally accepted that ob/ob mice display a dramatic increase in ß-cell mass to compensate for increased insulin demand, the spatial and quantitative dynamics of ß-cell mass distribution in this model has not been assessed by modern optical 3D imaging techniques. We applied optical projection tomography and ultramicroscopy imaging to extract information about individual islet ß-cell volumes throughout the volume of ob/ob pancreas between 4 and 52 weeks of age. Our data show that cystic lesions constitute a significant volume of the hyperplastic ob/ob islets. We propose that these lesions are formed by a mechanism involving extravasation of red blood cells/plasma due to increased islet vessel blood flow and vessel instability. Further, our data indicate that the primary lobular compartments of the ob/ob pancreas have different potentials for expanding their ß-cell population. Unawareness of the characteristics of ß-cell expansion in ob/ob mice presented in this report may significantly influence ex vivo and in vivo assessments of this model in studies of ß-cell adaptation and function.


Subject(s)
Islets of Langerhans/diagnostic imaging , Islets of Langerhans/pathology , Pancreas/diagnostic imaging , Animals , Fibrin/metabolism , Hypertrophy , Imaging, Three-Dimensional , Mice, Obese , Pancreas/cytology , Pancreas/pathology , Tomography, Optical/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...