25 research outputs found

    Distribution and connectivity of messenger molecules in the control of energy metabolism : focus on neuropeptides and calcium binding proteins

    Get PDF
    Feeding is an essential and complex behavior which aims to provide the energy required for maintaining physiological homeostasis. The drive to feed is a powerful stimulus arising from metabolic demands, and reinforced by evolutionary pressure. The current epidemic in obesity, and associated disorders such as diabetes, makes it clinically vital to understand the mechanisms behind the control of energy metabolism. Feeding is a process governed by the central nervous system (CNS); particularly through the interplay between different hypothalamic nuclei. At the heart of the feeding neuro-circuitry lies the arcuate nucleus (ARC) which acts as a metabolic sensor, taking stock of the supply and demands of energy in the body, and coordinating food intake and energy expenditure. The work in this thesis aimed to explore the neuro-anatomical substrate of metabolic control, and the mediators involved. The ARC contains two distinct sets of functionally antagonistic neurons. One group of neurons express the orexigenic peptides, neuropeptide Y (NPY) and agouti gene related peptide (AGRP); while the other set expresses the anorexigenic peptides, proopiomelanocortin (POMC) and cocaine- and amphetamine-regulated transcript. In paper V, we describe the histochemistry of NPY/AGRP and POMC neurons with regard to their anatomical interrelationship at the cell body and terminal level. A common experimental problem is that the ARC NPY cell bodies are difficult to distinguish and visualize in electrophysiological experiments and for immunohistochemistry. Therefore, in paper III, a novel transgenic mouse which expresses bright Renilla green fluorescent protein in NPY neurons was generated. Using this model, a comprehensive map of NPY-expressing cells in the CNS was generated and the effects of the satiety-inducing gut-brain bombesin peptides on ARC neurons were explored. Bombesin was found to exert powerful depolarizing actions on NPY and POMC neurons alike. Calcium binding proteins (CaBPs) have been used extensively to delineate neuronal populations, but the ARC has not yet been subjected to such analysis. In Paper IV we show that three major CaBPs (calbindin D-28k, calretinin, and parvalbumin) are all expressed in the ARC, but displayed little co-localization with previously described cell groups. One exception was POMC neurons, of which distinct subpopulations stained for calbindin D-28k and calretinin, respectively. Another CaBP, nucleobindin 2 (NUCB2; also known as nesfatin), has recently been proposed as a central anorexigenic mediator. In Paper I, the CNS distribution of this protein was shown to include nuclei that participate in all three output channels of metabolic control, i.e. behavioral, endocrine and autonomic modulation. Our data also suggest that NUCB2 may not act as a cleaved and secreted messenger as proposed, but rather may play an intracellular role. The wide distribution of NUCB2 in the neuroendocrine system prompted us to explore this protein in the pancreas (Paper II). We show that NUCB2 is exclusively expressed in insulin-producing β cells, and that islet NUCB2 is dramatically decreased in the diabetic Goto-Kakizaki rat, an effect that is normalized by fasting. These data indicate that NUCB2 may play a role in metabolic control also outside of the CNS

    Genome‐wide CRISPR screen identifies ZIC2 as an essential gene that controls the cell fate of early mesodermal precursors to human heart progenitors

    Get PDF
    Cardiac progenitor formation is one of the earliest committed steps of human cardiogenesis and requires the cooperation of multiple gene sets governed by developmental signaling cascades. To determine the key regulators for cardiac progenitor formation, we have developed a two-stage genome-wide CRISPR-knockout screen. We mimicked the progenitor formation process by differentiating human pluripotent stem cells (hPSCs) into cardiomyocytes, monitored by two distinct stage markers of early cardiac mesodermal formation and commitment to a multipotent heart progenitor cell fate: MESP1 and ISL1, respectively. From the screen output, we compiled a list of 15 candidate genes. After validating seven of them, we identified ZIC2 as an essential gene for cardiac progenitor formation. ZIC2 is known as a master regulator of neurogenesis. hPSCs with ZIC2 mutated still express pluripotency markers. However, their ability to differentiate into cardiomyocytes was greatly attenuated. RNASeq profiling of the ZIC2-mutant cells revealed that the mutants switched their cell fate alternatively to the noncardiac cell lineage. Further, single cell RNA-seq analysis showed the ZIC2 mutants affected the apelin receptor-related signaling pathway during mesoderm formation. Our results provide a new link between ZIC2 and human cardiogenesis and document the potential power of a genome-wide unbiased CRISPR-knockout screen to identify the key steps in human mesoderm precursor cell- and heart progenitor cell-fate determination during in vitro hPSC cardiogenesis.Swedish Research Council for Health, Working Life and Welfare (Forte)Knut and Alice Wallenberg Foundation, KAW 2013.0028Swedish Research Council, 541-2013-8351, 539‐2013‐7002European Research Council Advanced Research Grant Award, AdG743225Publishe

    Human ISL1+ ventricular progenitors self-assemble into an in vivo functional heart patch and preserve cardiac function post infarction

    Get PDF
    The generation of human pluripotent stem cell (hPSC)-derived ventricular progenitors and their assembly into a 3-dimensional in vivo functional ventricular heart patch has remained an elusive goal. Herein, we report the generation of an enriched pool of hPSC-derived ventricular progenitors (HVPs), which can expand, differentiate, self-assemble, and mature into a functional ventricular patch in vivo without the aid of any gel or matrix. We documented a specific temporal window, in which the HVPs will engraft in vivo. On day 6 of differentiation, HVPs were enriched by depleting cells positive for pluripotency marker TRA-1-60 with magnetic-activated cell sorting (MACS), and 3 million sorted cells were sub-capsularly transplanted onto kidneys of NSG mice where, after 2 months, they formed a 7 mm x 3 mm x 4 mm myocardial patch resembling the ventricular wall. The graft acquired several features of maturation: expression of ventricular marker (MLC2v), desmosomes, appearance of T-tubule-like structures, and electrophysiological action potential signature consistent with maturation, all this in a non-cardiac environment. We further demonstrated that HVPs transplanted into un-injured hearts of NSG mice remain viable for up to 8 months. Moreover, transplantation of 2 million HVPs largely preserved myocardial contractile function following myocardial infarction. Taken together, our study reaffirms the promising idea of using progenitor cells for regenerative therapy.ERC AdG743225Swedish Research Council Distinguished Professor Grant Dnr 541-2013-8351The Knut and Alice Wallenberg Foundation (KAW Dnr 2013.0028)Horizon 2020 research and innovation programme grant agreement No 647714Publishe

    Classical Human Leukocyte Antigen Alleles and C4 Haplotypes Are Not Significantly Associated With Depression

    Get PDF
    Background The prevalence of depression is higher in individuals with autoimmune diseases, but the mechanisms underlying the observed comorbidities are unknown. Shared genetic etiology is a plausible explanation for the overlap, and in this study we tested whether genetic variation in the major histocompatibility complex (MHC), which is associated with risk for autoimmune diseases, is also associated with risk for depression. Methods We fine-mapped the classical MHC (chr6: 29.6–33.1 Mb), imputing 216 human leukocyte antigen (HLA) alleles and 4 complement component 4 (C4) haplotypes in studies from the Psychiatric Genomics Consortium Major Depressive Disorder Working Group and the UK Biobank. The total sample size was 45,149 depression cases and 86,698 controls. We tested for association between depression status and imputed MHC variants, applying both a region-wide significance threshold (3.9 × 10−6) and a candidate threshold (1.6 × 10−4). Results No HLA alleles or C4 haplotypes were associated with depression at the region-wide threshold. HLA-B*08:01 was associated with modest protection for depression at the candidate threshold for testing in HLA genes in the meta-analysis (odds ratio = 0.98, 95 confidence interval = 0.97–0.99). Conclusions We found no evidence that an increased risk for depression was conferred by HLA alleles, which play a major role in the genetic susceptibility to autoimmune diseases, or C4 haplotypes, which are strongly associated with schizophrenia. These results suggest that any HLA or C4 variants associated with depression either are rare or have very modest effect sizes

    The nesfatin-1 anorectic neural pathway and its role in stress, circulation, and reproduction

    No full text

    Blocking phospholamban with VHH intrabodies enhances contractility and relaxation in heart failure

    Get PDF
    The dysregulated physical interaction between two intracellular membrane proteins, the sarco/endoplasmic reticulum Ca2+ ATPase and its reversible inhibitor phospholamban, induces heart failure by inhibiting calcium cycling. While phospholamban is a bona-fide therapeutic target, approaches to selectively inhibit this protein remain elusive. Here, we report the in vivo application of intracellular acting antibodies (intrabodies), derived from the variable domain of camelid heavy-chain antibodies, to modulate the function of phospholamban. Using a synthetic VHH phage-display library, we identify intrabodies with high affinity and specificity for different conformational states of phospholamban. Rapid phenotypic screening, via modified mRNA transfection of primary cells and tissue, efficiently identifies the intrabody with most desirable features. Adeno-associated virus mediated delivery of this intrabody results in improvement of cardiac performance in a murine heart failure model. Our strategy for generating intrabodies to investigate cardiac disease combined with modified mRNA and adeno-associated virus screening could reveal unique future therapeutic opportunities.European Research Council (ERC) AdG 745225Swedish Research Council 541-2013-8351Publishe

    Weight Perturbation Alters Leptin Signal Transduction in a Region-Specific Manner throughout the Brain.

    Get PDF
    Diet-induced obesity (DIO) resulting from consumption of a high fat diet (HFD) attenuates normal neuronal responses to leptin and may contribute to the metabolic defense of an acquired higher body weight in humans; the molecular bases for the persistence of this defense are unknown. We measured the responses of 23 brain regions to exogenous leptin in 4 different groups of weight- and/or diet-perturbed mice. Responses to leptin were assessed by quantifying pSTAT3 levels in brain nuclei 30 minutes following 3 mg/kg intraperitoneal leptin. HFD attenuated leptin sensing throughout the brain, but weight loss did not restore central leptin signaling to control levels in several brain regions important in energy homeostasis, including the arcuate and dorsomedial hypothalamic nuclei. Effects of diet on leptin signaling varied by brain region, with results dependent on the method of weight loss (restriction of calories of HFD, ad lib intake of standard mouse chow). High fat diet attenuates leptin signaling throughout the brain, but some brain regions maintain their ability to sense leptin. Weight loss restores leptin sensing to some degree in most (but not all) brain regions, while other brain regions display hypersensitivity to leptin following weight loss. Normal leptin sensing was restored in several brain regions, with the pattern of restoration dependent on the method of weight loss

    Human ISL1+ ventricular progenitors self-assemble into an in vivo functional heart patch and preserve cardiac function post infarction

    No full text
    The generation of human pluripotent stem cell (hPSC)-derived ventricular progenitors and their assembly into a 3-dimensional in vivo functional ventricular heart patch has remained an elusive goal. Herein, we report the generation of an enriched pool of hPSC-derived ventricular progenitors (HVPs), which can expand, differentiate, self-assemble, and mature into a functional ventricular patch in vivo without the aid of any gel or matrix. We documented a specific temporal window, in which the HVPs will engraft in vivo. On day 6 of differentiation, HVPs were enriched by depleting cells positive for pluripotency marker TRA-1-60 with magnetic-activated cell sorting (MACS), and 3 million sorted cells were sub-capsularly transplanted onto kidneys of NSG mice where, after 2 months, they formed a 7 mm × 3 mm × 4 mm myocardial patch resembling the ventricular wall. The graft acquired several features of maturation: expression of ventricular marker (MLC2v), desmosomes, appearance of T-tubule-like structures, and electrophysiological action potential signature consistent with maturation, all this in a non-cardiac environment. We further demonstrated that HVPs transplanted into un-injured hearts of NSG mice remain viable for up to 8 months. Moreover, transplantation of 2 million HVPs largely preserved myocardial contractile function following myocardial infarction. Taken together, our study reaffirms the promising idea of using progenitor cells for regenerative therapy
    corecore