1
|
Scully DM, Xia T, Musina GR, McCown MA, Umezu K, Kircher BK, Behringer RR, Larina IV. Region-specific roles of oviductal motile cilia in oocyte/embryo transport and fertility†. Biol Reprod 2025; 112:651-662. [PMID: 39761349 PMCID: PMC11996758 DOI: 10.1093/biolre/ioaf005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 12/18/2024] [Accepted: 01/05/2025] [Indexed: 03/29/2025] Open
Abstract
The physiological and clinical importance of motile cilia in reproduction is well recognized; however, the specific role they play in transport through the oviduct and how ciliopathies lead to subfertility and infertility are still unclear. The contribution of cilia beating, fluid flow, and smooth muscle contraction to overall progressive transport within the oviduct remains under debate. Therefore, we investigated the role of cilia in the oviduct transport of preimplantation eggs/embryos using a combination of genetic and advanced imaging approaches. We show that the region of the oviduct where cumulus-oocyte complex circling occurs, around the time of fertilization, is correlated with asymmetrical mucosal fold arrangement and non-radially distributed ciliated epithelium. Our results suggest that motile cilia, as well as mucosal fold asymmetry, may contribute to the local flow fields that help steer luminal contents away from the epithelial walls. We also present, in vivo, volumetric evidence of delayed egg transport in a genetic mouse model with disrupted motile cilia function in the female reproductive system. Females with Dnah5 deleted in the oviduct epithelium are subfertile and demonstrate disrupted motile cilia activity within the oviduct mucosa. Fifty percent of Dnah5 mutant females have delayed egg transport where cumulus-oocyte complexes did not progress to the ampulla at the expected time point and remained within the ovarian bursa. The integration of advanced imaging with genetic dysfunction of motile cilia provides valuable insights into oviductal transport. Potentially, these data could be valuable for better understanding and management of tubal pathologies and human infertility.
Collapse
Affiliation(s)
- Deirdre M Scully
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Tian Xia
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Guzel R Musina
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA
| | - Michaela A McCown
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Kohei Umezu
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Bonnie K Kircher
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Richard R Behringer
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Irina V Larina
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
2
|
Tan J, Duron A, Sucov HM, Makita T. Placode and neural crest origins of congenital deafness in mouse models of Waardenburg-Shah syndrome. iScience 2025; 28:111680. [PMID: 39868048 PMCID: PMC11762213 DOI: 10.1016/j.isci.2024.111680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 07/19/2024] [Accepted: 12/20/2024] [Indexed: 01/28/2025] Open
Abstract
Mutations in the human genes encoding the endothelin ligand-receptor pair EDN3 and EDNRB cause Waardenburg-Shah syndrome (WS4), which includes congenital hearing impairment. The current explanation for auditory dysfunction is defective migration of neural crest-derived melanocytes to the inner ear. We explored the role of endothelin signaling in auditory development in mice using neural crest-specific and placode-specific Ednrb mutation plus related genetic resources. On an outbred strain background, we find a normal representation of melanocytes in hearing-impaired mutant mice. Instead, our results in neural crest-specific Ednrb mutants implicate a previously unrecognized role for glial support of synapse assembly between auditory neurons and cochlear hair cells. Placode-specific Ednrb mutation also caused impaired hearing, resulting from deficient synaptic transmission. Our observations demonstrate the significant influence of genetic modifiers in auditory development, and invoke independent and separable roles for endothelin signaling in the neural crest and placode lineages to create a functional auditory circuitry.
Collapse
Affiliation(s)
- Jaime Tan
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Alicia Duron
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Henry M. Sucov
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Takako Makita
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
3
|
Coren L, Zaffryar-Eilot S, Odeh A, Kaganovsky A, Hasson P. Fibroblast diversification is an embryonic process dependent on muscle contraction. Cell Rep 2024; 43:115034. [PMID: 39636726 DOI: 10.1016/j.celrep.2024.115034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 09/18/2024] [Accepted: 11/14/2024] [Indexed: 12/07/2024] Open
Abstract
Fibroblasts, the most common cell type found in connective tissues, play major roles in development, homeostasis, regeneration, and disease. Although specific fibroblast subpopulations have been associated with different biological processes, the mechanisms and unique activities underlying their diversity have not been thoroughly examined. Here, we set out to dissect the variation in skeletal-muscle-resident fibroblasts (mrFibroblasts) during development. Our results demonstrate that mrFibroblasts diversify following the transition from embryonic to fetal myogenesis prior to birth. We find that mrFibroblasts segregate into two major subpopulations occupying distinct niches, with interstitial fibroblasts residing between the muscle fibers and delineating fibroblasts sheathing the muscle. We further show that these subpopulations entail distinct cellular dynamics and transcriptomes. Notably, we find that mrFibroblast subpopulations exert distinct regulatory roles on myoblast proliferation and differentiation. Finally, we demonstrate that this diversification depends on muscle contraction. Altogether, these findings establish that mrFibroblasts diversify in a spatiotemporal embryonic process into distinct cell types, entailing different characteristics and roles.
Collapse
Affiliation(s)
- Lavi Coren
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Shelly Zaffryar-Eilot
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Anas Odeh
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Anna Kaganovsky
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Peleg Hasson
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa 31096, Israel.
| |
Collapse
|
4
|
Poltavski DM, Cunha AT, Tan J, Sucov HM, Makita T. Lineage-specific intersection of endothelin and GDNF signaling in enteric nervous system development. eLife 2024; 13:RP96424. [PMID: 39641974 PMCID: PMC11623925 DOI: 10.7554/elife.96424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024] Open
Abstract
Two major ligand-receptor signaling axes - endothelin Edn3 and its receptor Ednrb, and glial-derived neurotrophic factor (GDNF) and its receptor Ret - are required for migration of enteric nervous system (ENS) progenitors to the hindgut. Mutations in either component cause colonic aganglionosis, also called Hirschsprung disease. Here, we have used Wnt1Cre and Pax2Cre in mice to show that these driver lines label distinct ENS lineages during progenitor migration and in their terminal hindgut fates. Both Cre lines result in Hirschsprung disease when combined with conditional Ednrb or conditional Ret alleles. In vitro explant assays and analysis of lineage-labeled mutant embryos show that GDNF but not Edn3 is a migration cue for cells of both lineages. Instead, Edn3-Ednrb function is required in both for GDNF responsiveness albeit in different ways: by expanding the Ret+ population in the Pax2Cre lineage, and by supporting Ret function in Wnt1Cre-derived cells. Our results demonstrate that two distinct lineages of progenitors give rise to the ENS, and that these divergently utilize endothelin signaling to support migration to the hindgut.
Collapse
Affiliation(s)
- Denise M Poltavski
- Department of Regenerative Medicine and Cell Biology, Medical University of South CarolinaCharlestonUnited States
| | - Alexander T Cunha
- Department of Regenerative Medicine and Cell Biology, Medical University of South CarolinaCharlestonUnited States
| | - Jaime Tan
- Department of Regenerative Medicine and Cell Biology, Medical University of South CarolinaCharlestonUnited States
| | - Henry M Sucov
- Department of Regenerative Medicine and Cell Biology, Medical University of South CarolinaCharlestonUnited States
| | - Takako Makita
- Department of Regenerative Medicine and Cell Biology, Medical University of South CarolinaCharlestonUnited States
| |
Collapse
|
5
|
Kim PH, Kim JR, Heizer PJ, Jung H, Tu Y, Presnell A, Scheithauer J, Yu RG, Young SG, Fong LG. The Accumulation of Progerin Underlies the Loss of Aortic Smooth Muscle Cells in Hutchinson-Gilford Progeria Syndrome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.29.620896. [PMID: 39554077 PMCID: PMC11565845 DOI: 10.1101/2024.10.29.620896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a progeroid disorder characterized by multiple aging-like phenotypes, including disease in large arteries. HGPS is caused by an internally truncated prelamin A (progerin) that cannot undergo the ZMPSTE24-mediated processing step that converts farnesyl-prelamin A to mature lamin A; consequently, progerin retains a carboxyl-terminal farnesyl lipid anchor. In cultured cells, progerin and full-length farnesyl-prelamin A (produced in Zmpste24 -/- cells) form an abnormal nuclear lamin meshwork accompanied by nuclear membrane ruptures and cell death; however, these proteins differ in their capacity to cause arterial disease. In a mouse model of HGPS (Lmna G609G), progerin causes loss of aortic smooth muscle cells (SMCs) by ~12 weeks of age. In contrast, farnesyl-prelamin A in Zmpste24 -/- mice does not cause SMC loss-even at 21 weeks of age. In young mice, aortic levels of farnesyl-prelamin A in Zmpste24 -/- mice and aortic levels of progerin in Lmna G609G/+ mice are the same. However, the levels of progerin and other A-type lamins increase with age in Lmna G609G/+ mice, whereas farnesyl-prelamin A and lamin C levels in Zmpste24 -/- mice remain stable. Lmna transcript levels are similar, implying that progerin influences nuclear lamin turnover. We identified a likely mechanism. In cultured SMCs, the phosphorylation of Ser-404 by AKT (which triggers prelamin A degradation) is reduced in progerin. In mice, AKT activity is significantly lower in Lmna G609G/+ aortas than in wild-type or Zmpste24 -/- aortas. Our studies identify that the accumulation of progerin in Lmna G609G aortas underlies the hallmark arterial pathology in HGPS.
Collapse
Affiliation(s)
- Paul H. Kim
- Department of Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Joonyoung R. Kim
- Department of Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Patrick J. Heizer
- Department of Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Hyesoo Jung
- Department of Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Yiping Tu
- Department of Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Ashley Presnell
- Department of Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Julia Scheithauer
- Department of Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Rachel G. Yu
- Department of Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Stephen G. Young
- Department of Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
- Department of Human Genetics, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Loren G. Fong
- Department of Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
| |
Collapse
|
6
|
Ozturk T, Mignot J, Gattazzo F, Gervais M, Relaix F, Rouard H, Didier N. Dual inhibition of P38 MAPK and JNK pathways preserves stemness markers and alleviates premature activation of muscle stem cells during isolation. Stem Cell Res Ther 2024; 15:179. [PMID: 38902774 PMCID: PMC11191274 DOI: 10.1186/s13287-024-03795-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 06/08/2024] [Indexed: 06/22/2024] Open
Abstract
BACKGROUND Adult skeletal muscle contains resident muscle stem cells (MuSC) with high myogenic and engraftment potentials, making them suitable for cell therapy and regenerative medicine approaches. However, purification process of MuSC remains a major hurdle to their use in the clinic. Indeed, muscle tissue enzymatic dissociation triggers a massive activation of stress signaling pathways, among which P38 and JNK MAPK, associated with a premature loss of MuSC quiescence. While the role of these pathways in the myogenic progression of MuSC is well established, the extent to which their dissociation-induced activation affects the functionality of these cells remains unexplored. METHODS We assessed the effect of P38 and JNK MAPK induction on stemness marker expression and MuSC activation state during isolation by pharmacological approaches. MuSC functionality was evaluated by in vitro assays and in vivo transplantation experiments. We performed a comparative analysis of the transcriptome of human MuSC purified with pharmacological inhibitors of P38 and JNK MAPK (SB202190 and SP600125, respectively) versus available RNAseq resources. RESULTS We monitored PAX7 protein levels in murine MuSC during muscle dissociation and demonstrated a two-step decline partly dependent on P38 and JNK MAPK activities. We showed that simultaneous inhibition of these pathways throughout the MuSC isolation process preserves the expression of stemness markers and limits their premature activation, leading to improved survival and amplification in vitro as well as increased engraftment in vivo. Through a comparative RNAseq analysis of freshly isolated human MuSC, we provide evidence that our findings in murine MuSC could be relevant to human MuSC. Based on these findings, we implemented a purification strategy, significantly improving the recovery yields of human MuSC. CONCLUSION Our study highlights the pharmacological limitation of P38 and JNK MAPK activities as a suitable strategy to qualitatively and quantitatively ameliorate human MuSC purification process, which could be of great interest for cell-based therapies.
Collapse
Affiliation(s)
- Teoman Ozturk
- Univ Paris Est Creteil, INSERM, EFS, IMRB, 94010, Creteil, France
| | - Julien Mignot
- Univ Paris Est Creteil, INSERM, EFS, IMRB, 94010, Creteil, France
| | | | - Marianne Gervais
- Univ Paris Est Creteil, INSERM, EFS, IMRB, 94010, Creteil, France
| | - Frédéric Relaix
- Univ Paris Est Creteil, INSERM, EFS, IMRB, 94010, Creteil, France
- EnvA, IMRB, 94700, Maisons-Alfort, France
- AP-HP, Hopital Mondor, Service d'histologie, 94010, Creteil, France
| | - Hélène Rouard
- Univ Paris Est Creteil, INSERM, EFS, IMRB, 94010, Creteil, France
- AP-HP, Hopital Mondor, Service d'histologie, 94010, Creteil, France
| | - Nathalie Didier
- Univ Paris Est Creteil, INSERM, EFS, IMRB, 94010, Creteil, France.
| |
Collapse
|
7
|
Shakarchy A, Zarfati G, Hazak A, Mealem R, Huk K, Ziv T, Avinoam O, Zaritsky A. Machine learning inference of continuous single-cell state transitions during myoblast differentiation and fusion. Mol Syst Biol 2024; 20:217-241. [PMID: 38238594 PMCID: PMC10912675 DOI: 10.1038/s44320-024-00010-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 12/19/2023] [Accepted: 01/04/2024] [Indexed: 03/06/2024] Open
Abstract
Cells modify their internal organization during continuous state transitions, supporting functions from cell division to differentiation. However, tools to measure dynamic physiological states of individual transitioning cells are lacking. We combined live-cell imaging and machine learning to monitor ERK1/2-inhibited primary murine skeletal muscle precursor cells, that transition rapidly and robustly from proliferating myoblasts to post-mitotic myocytes and then fuse, forming multinucleated myotubes. Our models, trained using motility or actin intensity features from single-cell tracking data, effectively tracked real-time continuous differentiation, revealing that differentiation occurs 7.5-14.5 h post induction, followed by fusion ~3 h later. Co-inhibition of ERK1/2 and p38 led to differentiation without fusion. Our model inferred co-inhibition leads to terminal differentiation, indicating that p38 is specifically required for transitioning from terminal differentiation to fusion. Our model also predicted that co-inhibition leads to changes in actin dynamics. Mass spectrometry supported these in silico predictions and suggested novel fusion and maturation regulators downstream of differentiation. Collectively, this approach can be adapted to various biological processes to uncover novel links between dynamic single-cell states and their functional outcomes.
Collapse
Affiliation(s)
- Amit Shakarchy
- Department of Software and Information Systems Engineering, Ben-Gurion University of the Negev, Beer-Sheva, 84105, Israel
| | - Giulia Zarfati
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, 761001, Israel
| | - Adi Hazak
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, 761001, Israel
| | - Reut Mealem
- Department of Software and Information Systems Engineering, Ben-Gurion University of the Negev, Beer-Sheva, 84105, Israel
| | - Karina Huk
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, 761001, Israel
| | - Tamar Ziv
- The Smoler Proteomics Center, Lorry I. Lokey Interdisciplinary Center for Life Sciences and Engineering, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Ori Avinoam
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, 761001, Israel.
| | - Assaf Zaritsky
- Department of Software and Information Systems Engineering, Ben-Gurion University of the Negev, Beer-Sheva, 84105, Israel.
| |
Collapse
|
8
|
Khatif H, Bazzi H. Generation and characterization of a Dkk4-Cre knock-in mouse line. Genesis 2024; 62:e23532. [PMID: 37435631 DOI: 10.1002/dvg.23532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 06/16/2023] [Accepted: 06/19/2023] [Indexed: 07/13/2023]
Abstract
Ectodermal appendages in mammals, such as teeth, mammary glands, sweat glands and hair follicles, are generated during embryogenesis through a series of mesenchymal-epithelial interactions. Canonical Wnt signaling and its inhibitors are implicated in the early steps of ectodermal appendage development and patterning. To study the activation dynamics of the Wnt target and inhibitor Dickkopf4 (Dkk4) in ectodermal appendages, we used CRSIPR/Cas9 to generate a Dkk4-Cre knock-in mouse (Mus musculus) line, where the Cre recombinase cDNA replaces the expression of endogenous Dkk4. Using Cre reporters, the Dkk4-Cre activity was evident at the prospective sites of ectodermal appendages, overlapping with the Dkk4 mRNA expression. Unexpectedly, a predominantly mesenchymal cell population in the embryo posterior also showed Dkk4-Cre activity. Lineage-tracing suggested that these cells are likely derived from a few Dkk4-Cre-expressing cells in the epiblast at early gastrulation. Finally, our analyses of Dkk4-Cre-expressing cells in developing hair follicle epithelial placodes revealed intra- and inter-placodal cellular heterogeneity, supporting emerging data on the positional and transcriptional cellular variability in placodes. Collectively, we propose the new Dkk4-Cre knock-in mouse line as a suitable model to study Wnt and DKK4 inhibitor dynamics in early mouse development and ectodermal appendage morphogenesis.
Collapse
Affiliation(s)
- Houda Khatif
- Department of Dermatology and Venereology, University Hospital of Cologne, University of Cologne, Cologne, Germany
- The Cologne Cluster of Excellence in Cellular Stress Responses in Aging-associated Diseases (CECAD), Medical Faculty, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Medical Faculty, University of Cologne, Cologne, Germany
| | - Hisham Bazzi
- Department of Dermatology and Venereology, University Hospital of Cologne, University of Cologne, Cologne, Germany
- The Cologne Cluster of Excellence in Cellular Stress Responses in Aging-associated Diseases (CECAD), Medical Faculty, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Medical Faculty, University of Cologne, Cologne, Germany
| |
Collapse
|
9
|
Karpova Y, Orlicky DJ, Schmidt EE, Tulin AV. Disrupting Poly(ADP-ribosyl)ating Pathway Creates Premalignant Conditions in Mammalian Liver. Int J Mol Sci 2023; 24:17205. [PMID: 38139034 PMCID: PMC10743425 DOI: 10.3390/ijms242417205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/01/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a major global health concern, representing one of the leading causes of cancer-related deaths. Despite various treatment options, the prognosis for HCC patients remains poor, emphasizing the need for a deeper understanding of the factors contributing to HCC development. This study investigates the role of poly(ADP-ribosyl)ation in hepatocyte maturation and its impact on hepatobiliary carcinogenesis. A conditional Parg knockout mouse model was employed, utilizing Cre recombinase under the albumin promoter to target Parg depletion specifically in hepatocytes. The disruption of the poly(ADP-ribosyl)ating pathway in hepatocytes affects the early postnatal liver development. The inability of hepatocytes to finish the late maturation step that occurs early after birth causes intensive apoptosis and acute inflammation, resulting in hypertrophic liver tissue with enlarged hepatocytes. Regeneration nodes with proliferative hepatocytes eventually replace the liver tissue and successfully fulfill the liver function. However, early developmental changes predispose these types of liver to develop pathologies, including with a malignant nature, later in life. In a chemically induced liver cancer model, Parg-depleted livers displayed a higher tendency for hepatocellular carcinoma development. This study underscores the critical role of the poly(ADP-ribosyl)ating pathway in hepatocyte maturation and highlights its involvement in liver pathologies and hepatobiliary carcinogenesis. Understanding these processes may provide valuable insights into liver biology and liver-related diseases, including cancer.
Collapse
Affiliation(s)
- Yaroslava Karpova
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, 501 North Columbia Road, Grand Forks, ND 58202, USA;
- Koltzov Institute of Developmental Biology of Russian Academy of Sciences, 119334 Moscow, Russia
| | - David J. Orlicky
- Department of Pathology, University of Colorado School of Medicine, Aurora, CO 80045, USA;
| | - Edward E. Schmidt
- Microbiology & Cell Biology, Montana State University, Bozeman, MT 59718, USA;
- Department of Microbiology & Immunology, Lewis Hall, Bozeman, MT 59718, USA
- Redox Biology Laboratory, University of Veterinary Medicine, 1078 Budapest, Hungary
| | - Alexei V. Tulin
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, 501 North Columbia Road, Grand Forks, ND 58202, USA;
| |
Collapse
|
10
|
Zhang AMY, Xia YH, Lin JSH, Chu KH, Wang WCK, Ruiter TJJ, Yang JCC, Chen N, Chhuor J, Patil S, Cen HH, Rideout EJ, Richard VR, Schaeffer DF, Zahedi RP, Borchers CH, Johnson JD, Kopp JL. Hyperinsulinemia acts via acinar insulin receptors to initiate pancreatic cancer by increasing digestive enzyme production and inflammation. Cell Metab 2023; 35:2119-2135.e5. [PMID: 37913768 DOI: 10.1016/j.cmet.2023.10.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 06/02/2023] [Accepted: 10/06/2023] [Indexed: 11/03/2023]
Abstract
The rising pancreatic cancer incidence due to obesity and type 2 diabetes is closely tied to hyperinsulinemia, an independent cancer risk factor. Previous studies demonstrated reducing insulin production suppressed pancreatic intraepithelial neoplasia (PanIN) pre-cancerous lesions in Kras-mutant mice. However, the pathophysiological and molecular mechanisms remained unknown, and in particular it was unclear whether hyperinsulinemia affected PanIN precursor cells directly or indirectly. Here, we demonstrate that insulin receptors (Insr) in KrasG12D-expressing pancreatic acinar cells are dispensable for glucose homeostasis but necessary for hyperinsulinemia-driven PanIN formation in the context of diet-induced hyperinsulinemia and obesity. Mechanistically, this was attributed to amplified digestive enzyme protein translation, triggering of local inflammation, and PanIN metaplasia in vivo. In vitro, insulin dose-dependently increased acinar-to-ductal metaplasia formation in a trypsin- and Insr-dependent manner. Collectively, our data shed light on the mechanisms connecting obesity-driven hyperinsulinemia and pancreatic cancer development.
Collapse
Affiliation(s)
- Anni M Y Zhang
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Yi Han Xia
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Jeffrey S H Lin
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Ken H Chu
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Wei Chuan K Wang
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Titine J J Ruiter
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Jenny C C Yang
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Nan Chen
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Justin Chhuor
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Shilpa Patil
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Haoning Howard Cen
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Elizabeth J Rideout
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Vincent R Richard
- Segal Cancer Proteomics Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada
| | - David F Schaeffer
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z7, Canada
| | - Rene P Zahedi
- Segal Cancer Proteomics Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada; Department of Internal Medicine, University of Manitoba, Winnipeg, MB R3A 1R9, Canada; Manitoba Centre for Proteomics and Systems Biology, Winnipeg, MB R3E 3P4, Canada
| | - Christoph H Borchers
- Segal Cancer Proteomics Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada; Gerald Bronfman Department of Oncology, Jewish General Hospital, McGill University, Montreal, QC H4A 3T2, Canada
| | - James D Johnson
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada.
| | - Janel L Kopp
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada.
| |
Collapse
|
11
|
Ozguldez HO, Govindasamy N, Fan R, Long H, Mildner K, Zeuschner D, Trappmann B, Ranga A, Bedzhov I. Polarity inversion reorganizes the stem cell compartment of the trophoblast lineage. Cell Rep 2023; 42:112313. [PMID: 36989113 PMCID: PMC10157138 DOI: 10.1016/j.celrep.2023.112313] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 01/12/2023] [Accepted: 03/13/2023] [Indexed: 03/30/2023] Open
Abstract
The extra-embryonic tissues that form the placenta originate from a small population of trophectoderm cells with stem cell properties, positioned at the embryonic pole of the mouse blastocyst. During the implantation stages, the polar trophectoderm rapidly proliferates and transforms into extra-embryonic ectoderm. The current model of trophoblast morphogenesis suggests that tissue folding reshapes the trophoblast during the blastocyst to egg cylinder transition. Instead of through folding, here we found that the tissue scale architecture of the stem cell compartment of the trophoblast lineage is reorganized via inversion of the epithelial polarity axis. Our findings show the developmental significance of polarity inversion and provide a framework for the morphogenetic transitions in the peri-implantation trophoblast.
Collapse
Affiliation(s)
- Hatice O Ozguldez
- Embryonic Self-Organization Research Group, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany
| | - Niraimathi Govindasamy
- Embryonic Self-Organization Research Group, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany
| | - Rui Fan
- Embryonic Self-Organization Research Group, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany
| | - Hongyan Long
- Bioactive Materials Laboratory, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany
| | - Karina Mildner
- Electron Microscopy Facility, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany
| | - Dagmar Zeuschner
- Electron Microscopy Facility, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany
| | - Britta Trappmann
- Bioactive Materials Laboratory, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany
| | - Adrian Ranga
- Laboratory of Bioengineering and Morphogenesis, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| | - Ivan Bedzhov
- Embryonic Self-Organization Research Group, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany.
| |
Collapse
|
12
|
Chai W, Hao W, Liu J, Han Z, Chang S, Cheng L, Sun M, Yan G, Liu Z, Liu Y, Zhang G, Xing L, Chen H, Liu P. Visualizing Cathepsin K-Cre Expression at the Single-Cell Level with GFP Reporters. JBMR Plus 2022; 7:e10706. [PMID: 36699636 PMCID: PMC9850439 DOI: 10.1002/jbm4.10706] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 11/10/2022] [Accepted: 11/16/2022] [Indexed: 11/23/2022] Open
Abstract
The Cre/lox system is a fundamental tool for functional genomic studies, and a number of Cre lines have been generated to target genes of interest spatially and temporally in defined cells or tissues; this approach has greatly expanded our knowledge of gene functions. However, the limitations of this system have recently been recognized, and we must address the challenge of so-called nonspecific/off-target effects when a Cre line is utilized to investigate a gene of interest. For example, cathepsin K (Ctsk) has been used as a specific osteoclast marker, and Cre driven by its promoter is widely utilized for osteoclast investigations. However, Ctsk-Cre expression has recently been identified in other cell types, such as osteocytes, periosteal stem cells, and tenocytes. To better understand Ctsk-Cre expression and ensure appropriate use of this Cre line, we performed a comprehensive analysis of Ctsk-Cre expression at the single-cell level in major organs and tissues using two green fluorescent protein (GFP) reporters (ROSA nT-nG and ROSA tdT) and a tissue clearing technique in young and aging mice. The expression profile was further verified by immunofluorescence staining and droplet digital RT-PCR. The results demonstrate that Ctsk-Cre is expressed not only in osteoclasts but also at various levels in osteoblast lineage cells and other major organs/tissues, particularly in the brain, kidney, pancreas, and blood vessels. Furthermore, Ctsk-Cre expression increases markedly in the bone marrow, skeletal muscle, and intervertebral discs in aging mice. These data will be valuable for accurately interpreting data obtained from in vivo studies using Ctsk-Cre mice to avoid potentially misleading conclusions. © 2022 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Wenhuan Chai
- Laboratory of Bone & Adipose BiologyShanxi Medical UniversityTaiyuanChina
| | - Weiwei Hao
- Laboratory of Bone & Adipose BiologyShanxi Medical UniversityTaiyuanChina
| | - Jintao Liu
- Laboratory of Bone & Adipose BiologyShanxi Medical UniversityTaiyuanChina
| | - Zhenglin Han
- Laboratory of Bone & Adipose BiologyShanxi Medical UniversityTaiyuanChina
| | - Shiyu Chang
- Laboratory of Bone & Adipose BiologyShanxi Medical UniversityTaiyuanChina
| | - Liben Cheng
- Laboratory of Bone & Adipose BiologyShanxi Medical UniversityTaiyuanChina
| | - Mingxin Sun
- Laboratory of Bone & Adipose BiologyShanxi Medical UniversityTaiyuanChina
| | - Guofang Yan
- Laboratory of Bone & Adipose BiologyShanxi Medical UniversityTaiyuanChina
| | - Zemin Liu
- Laboratory of Bone & Adipose BiologyShanxi Medical UniversityTaiyuanChina
| | - Yin Liu
- Laboratory of Bone & Adipose BiologyShanxi Medical UniversityTaiyuanChina
| | - Guodong Zhang
- Laboratory of Bone & Adipose BiologyShanxi Medical UniversityTaiyuanChina
| | - Li Xing
- Laboratory of Bone & Adipose BiologyShanxi Medical UniversityTaiyuanChina
| | - Hongqian Chen
- Laboratory of Bone & Adipose BiologyShanxi Medical UniversityTaiyuanChina
| | - Peng Liu
- Laboratory of Bone & Adipose BiologyShanxi Medical UniversityTaiyuanChina
| |
Collapse
|
13
|
Sefton EM, Gallardo M, Tobin CE, Collins BC, Colasanto MP, Merrell AJ, Kardon G. Fibroblast-derived Hgf controls recruitment and expansion of muscle during morphogenesis of the mammalian diaphragm. eLife 2022; 11:e74592. [PMID: 36154712 PMCID: PMC9514848 DOI: 10.7554/elife.74592] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 09/13/2022] [Indexed: 12/01/2022] Open
Abstract
The diaphragm is a domed muscle between the thorax and abdomen essential for breathing in mammals. Diaphragm development requires the coordinated development of muscle, connective tissue, and nerve, which are derived from different embryonic sources. Defects in diaphragm development cause the common and often lethal birth defect, congenital diaphragmatic hernias (CDH). HGF/MET signaling is required for diaphragm muscularization, but the source of HGF and the specific functions of this pathway in muscle progenitors and effects on phrenic nerve have not been explicitly tested. Using conditional mutagenesis in mice and pharmacological inhibition of MET, we demonstrate that the pleuroperitoneal folds (PPFs), transient embryonic structures that give rise to the connective tissue in the diaphragm, are the source of HGF critical for diaphragm muscularization. PPF-derived HGF is directly required for recruitment of MET+ muscle progenitors to the diaphragm and indirectly (via its effect on muscle development) required for phrenic nerve primary branching. In addition, HGF is continuously required for maintenance and motility of the pool of progenitors to enable full muscularization. Localization of HGF at the diaphragm's leading edges directs dorsal and ventral expansion of muscle and regulates its overall size and shape. Surprisingly, large muscleless regions in HGF and Met mutants do not lead to hernias. While these regions are likely more susceptible to CDH, muscle loss is not sufficient to cause CDH.
Collapse
Affiliation(s)
- Elizabeth M Sefton
- Department of Human Genetics, University of UtahSalt Lake CityUnited States
| | - Mirialys Gallardo
- Department of Human Genetics, University of UtahSalt Lake CityUnited States
| | - Claire E Tobin
- Department of Human Genetics, University of UtahSalt Lake CityUnited States
| | - Brittany C Collins
- Department of Human Genetics, University of UtahSalt Lake CityUnited States
| | - Mary P Colasanto
- Department of Human Genetics, University of UtahSalt Lake CityUnited States
| | | | - Gabrielle Kardon
- Department of Human Genetics, University of UtahSalt Lake CityUnited States
| |
Collapse
|
14
|
Teng T, Teng CS, Kaartinen V, Bush JO. A unique form of collective epithelial migration is crucial for tissue fusion in the secondary palate and can overcome loss of epithelial apoptosis. Development 2022; 149:275520. [PMID: 35593401 PMCID: PMC9188751 DOI: 10.1242/dev.200181] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 04/11/2022] [Indexed: 11/20/2022]
Abstract
Tissue fusion frequently requires the removal of an epithelium that intervenes distinct primordia to form one continuous structure. In the mammalian secondary palate, a midline epithelial seam (MES) forms between two palatal shelves and must be removed to allow mesenchymal confluence. Abundant apoptosis and cell extrusion support their importance in MES removal. However, genetically disrupting the intrinsic apoptotic regulators BAX and BAK within the MES results in complete loss of cell death and cell extrusion, but successful removal of the MES. Novel static- and live-imaging approaches reveal that the MES is removed through streaming migration of epithelial trails and islands to reach the oral and nasal epithelial surfaces. Epithelial trail cells that express the basal epithelial marker ΔNp63 begin to express periderm markers, suggesting that migration is concomitant with differentiation. Live imaging reveals anisotropic actomyosin contractility within epithelial trails, and genetic ablation of actomyosin contractility results in dispersion of epithelial collectives and failure of normal MES migration. These findings demonstrate redundancy between cellular mechanisms of morphogenesis, and reveal a crucial and unique form of collective epithelial migration during tissue fusion. Summary: Multiple cellular processes mediate secondary palate fusion, including a unique form of streaming collective epithelial migration driven by pulsatile actomyosin contractility.
Collapse
Affiliation(s)
- Teng Teng
- University of California San Francisco 1 Department of Cell and Tissue Biology , , San Francisco, CA 94143 , USA
- University of California San Francisco 2 Program in Craniofacial Biology , , San Francisco, CA 94143 , USA
- Institute for Human Genetics, University of California San Francisco 3 , San Francisco, CA 94143 , USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco 4 , San Francisco, CA 94143 , USA
| | - Camilla S. Teng
- University of California San Francisco 1 Department of Cell and Tissue Biology , , San Francisco, CA 94143 , USA
- University of California San Francisco 2 Program in Craniofacial Biology , , San Francisco, CA 94143 , USA
- Institute for Human Genetics, University of California San Francisco 3 , San Francisco, CA 94143 , USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco 4 , San Francisco, CA 94143 , USA
| | - Vesa Kaartinen
- University of Michigan School of Dentistry 5 Department of Biologic and Materials Sciences , , Ann Arbor, MI 48109 , USA
| | - Jeffrey O. Bush
- University of California San Francisco 1 Department of Cell and Tissue Biology , , San Francisco, CA 94143 , USA
- University of California San Francisco 2 Program in Craniofacial Biology , , San Francisco, CA 94143 , USA
- Institute for Human Genetics, University of California San Francisco 3 , San Francisco, CA 94143 , USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco 4 , San Francisco, CA 94143 , USA
| |
Collapse
|
15
|
Shen TH, Stauber J, Xu K, Jacunski A, Paragas N, Callahan M, Banlengchit R, Levitman AD, Desanti De Oliveira B, Beenken A, Grau MS, Mathieu E, Zhang Q, Li Y, Gopal T, Askanase N, Arumugam S, Mohan S, Good PI, Stevens JS, Lin F, Sia SK, Lin CS, D’Agati V, Kiryluk K, Tatonetti NP, Barasch J. Snapshots of nascent RNA reveal cell- and stimulus-specific responses to acute kidney injury. JCI Insight 2022; 7:e146374. [PMID: 35230973 PMCID: PMC8986083 DOI: 10.1172/jci.insight.146374] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The current strategy to detect acute injury of kidney tubular cells relies on changes in serum levels of creatinine. Yet serum creatinine (sCr) is a marker of both functional and pathological processes and does not adequately assay tubular injury. In addition, sCr may require days to reach diagnostic thresholds, yet tubular cells respond with programs of damage and repair within minutes or hours. To detect acute responses to clinically relevant stimuli, we created mice expressing Rosa26-floxed-stop uracil phosphoribosyltransferase (Uprt) and inoculated 4-thiouracil (4-TU) to tag nascent RNA at selected time points. Cre-driven 4-TU-tagged RNA was isolated from intact kidneys and demonstrated that volume depletion and ischemia induced different genetic programs in collecting ducts and intercalated cells. Even lineage-related cell types expressed different genes in response to the 2 stressors. TU tagging also demonstrated the transient nature of the responses. Because we placed Uprt in the ubiquitously active Rosa26 locus, nascent RNAs from many cell types can be tagged in vivo and their roles interrogated under various conditions. In short, 4-TU labeling identifies stimulus-specific, cell-specific, and time-dependent acute responses that are otherwise difficult to detect with other technologies and are entirely obscured when sCr is the sole metric of kidney damage.
Collapse
Affiliation(s)
| | | | | | - Alexandra Jacunski
- Department of Biomedical Informatics, Columbia University, New York, New York, USA
| | - Neal Paragas
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | - Sumit Mohan
- Department of Medicine, and
- Department of Epidemiology
| | | | | | | | | | - Chyuan-Sheng Lin
- Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | - Vivette D’Agati
- Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | | | | | | |
Collapse
|
16
|
Skovsø S, Panzhinskiy E, Kolic J, Cen HH, Dionne DA, Dai XQ, Sharma RB, Elghazi L, Ellis CE, Faulkner K, Marcil SAM, Overby P, Noursadeghi N, Hutchinson D, Hu X, Li H, Modi H, Wildi JS, Botezelli JD, Noh HL, Suk S, Gablaski B, Bautista A, Kim R, Cras-Méneur C, Flibotte S, Sinha S, Luciani DS, Nislow C, Rideout EJ, Cytrynbaum EN, Kim JK, Bernal-Mizrachi E, Alonso LC, MacDonald PE, Johnson JD. Beta-cell specific Insr deletion promotes insulin hypersecretion and improves glucose tolerance prior to global insulin resistance. Nat Commun 2022; 13:735. [PMID: 35136059 PMCID: PMC8826929 DOI: 10.1038/s41467-022-28039-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 01/05/2022] [Indexed: 01/23/2023] Open
Abstract
Insulin receptor (Insr) protein is present at higher levels in pancreatic β-cells than in most other tissues, but the consequences of β-cell insulin resistance remain enigmatic. Here, we use an Ins1cre knock-in allele to delete Insr specifically in β-cells of both female and male mice. We compare experimental mice to Ins1cre-containing littermate controls at multiple ages and on multiple diets. RNA-seq of purified recombined β-cells reveals transcriptomic consequences of Insr loss, which differ between female and male mice. Action potential and calcium oscillation frequencies are increased in Insr knockout β-cells from female, but not male mice, whereas only male βInsrKO islets have reduced ATP-coupled oxygen consumption rate and reduced expression of genes involved in ATP synthesis. Female βInsrKO and βInsrHET mice exhibit elevated insulin release in ex vivo perifusion experiments, during hyperglycemic clamps, and following i.p. glucose challenge. Deletion of Insr does not alter β-cell area up to 9 months of age, nor does it impair hyperglycemia-induced proliferation. Based on our data, we adapt a mathematical model to include β-cell insulin resistance, which predicts that β-cell Insr knockout improves glucose tolerance depending on the degree of whole-body insulin resistance. Indeed, glucose tolerance is significantly improved in female βInsrKO and βInsrHET mice compared to controls at 9, 21 and 39 weeks, and also in insulin-sensitive 4-week old males. We observe no improved glucose tolerance in older male mice or in high fat diet-fed mice, corroborating the prediction that global insulin resistance obscures the effects of β-cell specific insulin resistance. The propensity for hyperinsulinemia is associated with mildly reduced fasting glucose and increased body weight. We further validate our main in vivo findings using an Ins1-CreERT transgenic line and find that male mice have improved glucose tolerance 4 weeks after tamoxifen-mediated Insr deletion. Collectively, our data show that β-cell insulin resistance in the form of reduced β-cell Insr contributes to hyperinsulinemia in the context of glucose stimulation, thereby improving glucose homeostasis in otherwise insulin sensitive sex, dietary and age contexts.
Collapse
Affiliation(s)
- Søs Skovsø
- Diabetes Research Group, Life Sciences Institute, and Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Evgeniy Panzhinskiy
- Diabetes Research Group, Life Sciences Institute, and Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Jelena Kolic
- Diabetes Research Group, Life Sciences Institute, and Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Haoning Howard Cen
- Diabetes Research Group, Life Sciences Institute, and Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Derek A Dionne
- Diabetes Research Group, Life Sciences Institute, and Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Xiao-Qing Dai
- Alberta Diabetes Institute and Department of Pharmacology, University of Alberta, Edmonton, Canada
| | - Rohit B Sharma
- Division of Endocrinology, Diabetes and Metabolism and the Weill Center for Metabolic Health, Weill Cornell Medicine, New York, NY, USA
| | - Lynda Elghazi
- Department of Ophthalmology and Visual Sciences, University of Michigan Kellogg Eye Center, Ann Arbor, MI, USA
| | - Cara E Ellis
- Diabetes Research Group, Life Sciences Institute, and Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Katharine Faulkner
- Department of Mathematics, University of British Columbia, Vancouver, BC, Canada
| | - Stephanie A M Marcil
- Diabetes Research Group, Life Sciences Institute, and Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Peter Overby
- Diabetes Research Group, Life Sciences Institute, and Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Nilou Noursadeghi
- Diabetes Research Group, Life Sciences Institute, and Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Daria Hutchinson
- Diabetes Research Group, Life Sciences Institute, and Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Xiaoke Hu
- Diabetes Research Group, Life Sciences Institute, and Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Hong Li
- Diabetes Research Group, Life Sciences Institute, and Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Honey Modi
- Diabetes Research Group, Life Sciences Institute, and Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Jennifer S Wildi
- Diabetes Research Group, Life Sciences Institute, and Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - J Diego Botezelli
- Diabetes Research Group, Life Sciences Institute, and Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Hye Lim Noh
- Program in Molecular Medicine University of Massachusetts Medical School, Worcester, MA, USA
- Charles River Laboratories, Shrewsbury, MA, USA
| | - Sujin Suk
- Program in Molecular Medicine University of Massachusetts Medical School, Worcester, MA, USA
| | - Brian Gablaski
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
- Charles River Laboratories, Shrewsbury, MA, USA
| | - Austin Bautista
- Alberta Diabetes Institute and Department of Pharmacology, University of Alberta, Edmonton, Canada
| | - Ryekjang Kim
- Alberta Diabetes Institute and Department of Pharmacology, University of Alberta, Edmonton, Canada
| | - Corentin Cras-Méneur
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, MI, USA
| | - Stephane Flibotte
- UBC Life Sciences Institute Bioinformatics Facility, University of British Columbia, Vancouver, BC, Canada
| | - Sunita Sinha
- UBC Sequencing and Bioinformatics Consortium, Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Dan S Luciani
- BC Children's Hospital Research Institute, Department of Surgery, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Corey Nislow
- UBC Sequencing and Bioinformatics Consortium, Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Elizabeth J Rideout
- Diabetes Research Group, Life Sciences Institute, and Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Eric N Cytrynbaum
- Department of Mathematics, University of British Columbia, Vancouver, BC, Canada
| | - Jason K Kim
- Program in Molecular Medicine University of Massachusetts Medical School, Worcester, MA, USA
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Ernesto Bernal-Mizrachi
- Division of Endocrinology, Diabetes and Metabolism, University of Miami Miller School of Medicine and Miami VA Health Care System, Miami, FL, USA
| | - Laura C Alonso
- Division of Endocrinology, Diabetes and Metabolism and the Weill Center for Metabolic Health, Weill Cornell Medicine, New York, NY, USA
| | - Patrick E MacDonald
- Alberta Diabetes Institute and Department of Pharmacology, University of Alberta, Edmonton, Canada
| | - James D Johnson
- Diabetes Research Group, Life Sciences Institute, and Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
17
|
Experience-dependent myelination following stress is mediated by the neuropeptide dynorphin. Neuron 2021; 109:3619-3632.e5. [PMID: 34536353 DOI: 10.1016/j.neuron.2021.08.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 07/14/2021] [Accepted: 08/13/2021] [Indexed: 11/22/2022]
Abstract
Emerging evidence implicates experience-dependent myelination in learning and memory. However, the specific signals underlying this process remain unresolved. We demonstrate that the neuropeptide dynorphin, which is released from neurons upon high levels of activity, promotes experience-dependent myelination. Following forced swim stress, an experience that induces striatal dynorphin release, we observe increased striatal oligodendrocyte precursor cell (OPC) differentiation and myelination, which is abolished by deleting dynorphin or blocking its endogenous receptor, kappa opioid receptor (KOR). We find that dynorphin also promotes developmental OPC differentiation and myelination and demonstrate that this effect requires KOR expression specifically in OPCs. We characterize dynorphin-expressing neurons and use genetic sparse labeling to trace their axonal projections. Surprisingly, we find that they are unmyelinated normally and following forced swim stress. We propose a new model whereby experience-dependent and developmental myelination is mediated by unmyelinated, neuropeptide-expressing neurons that promote OPC differentiation for the myelination of neighboring axons.
Collapse
|
18
|
Yaseen W, Kraft-Sheleg O, Zaffryar-Eilot S, Melamed S, Sun C, Millay DP, Hasson P. Fibroblast fusion to the muscle fiber regulates myotendinous junction formation. Nat Commun 2021; 12:3852. [PMID: 34158500 PMCID: PMC8219707 DOI: 10.1038/s41467-021-24159-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 06/03/2021] [Indexed: 02/06/2023] Open
Abstract
Vertebrate muscles and tendons are derived from distinct embryonic origins yet they must interact in order to facilitate muscle contraction and body movements. How robust muscle tendon junctions (MTJs) form to be able to withstand contraction forces is still not understood. Using techniques at a single cell resolution we reexamine the classical view of distinct identities for the tissues composing the musculoskeletal system. We identify fibroblasts that have switched on a myogenic program and demonstrate these dual identity cells fuse into the developing muscle fibers along the MTJs facilitating the introduction of fibroblast-specific transcripts into the elongating myofibers. We suggest this mechanism resulting in a hybrid muscle fiber, primarily along the fiber tips, enables a smooth transition from muscle fiber characteristics towards tendon features essential for forming robust MTJs. We propose that dual characteristics of junctional cells could be a common mechanism for generating stable interactions between tissues throughout the musculoskeletal system.
Collapse
Affiliation(s)
- Wesal Yaseen
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa, Israel
| | - Ortal Kraft-Sheleg
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa, Israel
| | - Shelly Zaffryar-Eilot
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa, Israel
| | - Shay Melamed
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa, Israel
| | - Chengyi Sun
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Douglas P Millay
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Peleg Hasson
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
19
|
Nie X, Ricupero CL, Jiao K, Yang P, Mao JJ. mTOR deletion in neural crest cells disrupts cardiac outflow tract remodeling and causes a spectrum of cardiac defects through the mTORC1 pathway. Dev Biol 2021; 477:241-250. [PMID: 34052210 DOI: 10.1016/j.ydbio.2021.05.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 04/17/2021] [Accepted: 05/15/2021] [Indexed: 11/28/2022]
Abstract
A critical cell type participating in cardiac outflow tract development is a subpopulation of the neural crest cells, the cardiac neural crest cells (NCCs), whose defect causes a spectrum of cardiovascular abnormalities. Accumulating evidence indicates that mTOR, which belongs to the PI3K-related kinase family and impacts multiple signaling pathways in a variety of contexts, plays a pivotal role for NCC development. Here, we investigated functional roles of mTOR for cardiac neural crest development using several lines of mouse genetic models. We found that disruption of mTOR caused NCC defects and failure of cardiac outflow tract separation, which resulted in a spectrum of cardiac defects including persistent truncus arteriosus, ventricular septal defect and ventricular wall defect. Specifically, mutant neural crest cells showed reduced migration into the cardiac OFT and prematurely exited the cell cycle. A number of critical factors and fundamental signaling pathways, which are important for neural crest and cardiomyocyte development, were impaired. Moreover, actin dynamics was disrupted by mTOR deletion. Finally, by phenotyping the neural crest Rptor and Rictor knockout mice respectively, we demonstrate that mTOR acts principally through the mTORC1 pathway for cardiac neural crest cells. Altogether, these data established essential roles of mTOR for cardiac NCC development and imply that dysregulation of mTOR in NCCs may underline a spectrum of cardiac defects.
Collapse
Affiliation(s)
- Xuguang Nie
- Center for Birth Defects Research,Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, USA; College of Dental Medicine, Columbia University in the City of New York, New York, NY, USA.
| | - Christopher L Ricupero
- College of Dental Medicine, Columbia University in the City of New York, New York, NY, USA
| | - Kai Jiao
- University of Alabama at Birmingham, Department of Genetics and Genomic Sciences, Birmingham, AL, USA
| | - Peixin Yang
- Center for Birth Defects Research,Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jeremy J Mao
- College of Dental Medicine, Columbia University in the City of New York, New York, NY, USA.
| |
Collapse
|
20
|
Hoover C, Kondo Y, Shao B, McDaniel MJ, Lee R, McGee S, Whiteheart S, Bergmeier W, McEver RP, Xia L. Heightened activation of embryonic megakaryocytes causes aneurysms in the developing brain of mice lacking podoplanin. Blood 2021; 137:2756-2769. [PMID: 33619517 PMCID: PMC8138551 DOI: 10.1182/blood.2020010310] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 02/06/2021] [Indexed: 12/29/2022] Open
Abstract
During early embryonic development in mammals, including humans and mice, megakaryocytes (Mks) first originate from primitive hematopoiesis in the yolk sac. These embryonic Mks (eMks) circulate in the vasculature with unclear function. Herein, we report that podoplanin (PDPN), the ligand of C-type lectin-like receptor (CLEC-2) on Mks/platelets, is temporarily expressed in neural tissue during midgestation in mice. Loss of PDPN or CLEC-2 resulted in aneurysms and spontaneous hemorrhage, specifically in the lower diencephalon during midgestation. Surprisingly, more eMks/platelets had enhanced granule release and localized to the lower diencephalon in mutant mouse embryos than in wild-type littermates before hemorrhage. We found that PDPN counteracted the collagen-1-induced secretion of angiopoietin-1 from fetal Mks, which coincided with enhanced TIE-2 activation in aneurysm-like sprouts of PDPN-deficient embryos. Blocking platelet activation prevented the PDPN-deficient embryo from developing vascular defects. Our data reveal a new role for PDPN in regulating eMk function during midgestation.
Collapse
MESH Headings
- Aneurysm, Ruptured/embryology
- Aneurysm, Ruptured/etiology
- Angiopoietin-1/metabolism
- Animals
- Brain/blood supply
- Brain/embryology
- Cells, Cultured
- Cerebral Hemorrhage/embryology
- Cerebral Hemorrhage/etiology
- Collagen/pharmacology
- Diencephalon/blood supply
- Diencephalon/embryology
- Gene Expression Regulation, Developmental
- Gestational Age
- Intracranial Aneurysm/embryology
- Intracranial Aneurysm/etiology
- Intracranial Aneurysm/genetics
- Intracranial Aneurysm/pathology
- Lectins, C-Type/deficiency
- Lectins, C-Type/genetics
- Lectins, C-Type/physiology
- Megakaryocytes/metabolism
- Megakaryocytes/pathology
- Membrane Glycoproteins/deficiency
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/physiology
- Mice
- Mice, Knockout
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/physiopathology
- Neovascularization, Physiologic/physiology
- Platelet Activation
- Platelet Aggregation/drug effects
- Platelet Aggregation Inhibitors/pharmacology
- Receptor, TIE-2/metabolism
Collapse
Affiliation(s)
- Christopher Hoover
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Yuji Kondo
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Bojing Shao
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Michael J McDaniel
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Robert Lee
- Department of Biochemistry and Biophysics-UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC; and
| | - Samuel McGee
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Sidney Whiteheart
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY
| | - Wolfgang Bergmeier
- Department of Biochemistry and Biophysics-UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC; and
| | - Rodger P McEver
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Lijun Xia
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| |
Collapse
|
21
|
Blum JA, Klemm S, Shadrach JL, Guttenplan KA, Nakayama L, Kathiria A, Hoang PT, Gautier O, Kaltschmidt JA, Greenleaf WJ, Gitler AD. Single-cell transcriptomic analysis of the adult mouse spinal cord reveals molecular diversity of autonomic and skeletal motor neurons. Nat Neurosci 2021; 24:572-583. [PMID: 33589834 PMCID: PMC8016743 DOI: 10.1038/s41593-020-00795-0] [Citation(s) in RCA: 116] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Accepted: 12/23/2020] [Indexed: 01/30/2023]
Abstract
The spinal cord is a fascinating structure that is responsible for coordinating movement in vertebrates. Spinal motor neurons control muscle activity by transmitting signals from the spinal cord to diverse peripheral targets. In this study, we profiled 43,890 single-nucleus transcriptomes from the adult mouse spinal cord using fluorescence-activated nuclei sorting to enrich for motor neuron nuclei. We identified 16 sympathetic motor neuron clusters, which are distinguishable by spatial localization and expression of neuromodulatory signaling genes. We found surprising skeletal motor neuron heterogeneity in the adult spinal cord, including transcriptional differences that correlate with electrophysiologically and spatially distinct motor pools. We also provide evidence for a novel transcriptional subpopulation of skeletal motor neuron (γ*). Collectively, these data provide a single-cell transcriptional atlas ( http://spinalcordatlas.org ) for investigating the organizing molecular logic of adult motor neuron diversity, as well as the cellular and molecular basis of motor neuron function in health and disease.
Collapse
Affiliation(s)
- Jacob A Blum
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Neurosciences Graduate Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Sandy Klemm
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Jennifer L Shadrach
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Kevin A Guttenplan
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Neurosciences Graduate Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Lisa Nakayama
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Arwa Kathiria
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Phuong T Hoang
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Neurology and Neurological Science, Stanford University School of Medicine, Stanford, CA, USA
| | - Olivia Gautier
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Neurosciences Graduate Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Julia A Kaltschmidt
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - William J Greenleaf
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Applied Physics, Stanford University, Stanford, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Aaron D Gitler
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
22
|
Tyser RCV, Ibarra-Soria X, McDole K, Arcot Jayaram S, Godwin J, van den Brand TAH, Miranda AMA, Scialdone A, Keller PJ, Marioni JC, Srinivas S. Characterization of a common progenitor pool of the epicardium and myocardium. Science 2021; 371:eabb2986. [PMID: 33414188 PMCID: PMC7615359 DOI: 10.1126/science.abb2986] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 08/13/2020] [Accepted: 12/17/2020] [Indexed: 12/14/2022]
Abstract
The mammalian heart is derived from multiple cell lineages; however, our understanding of when and how the diverse cardiac cell types arise is limited. We mapped the origin of the embryonic mouse heart at single-cell resolution using a combination of transcriptomic, imaging, and genetic lineage labeling approaches. This mapping provided a transcriptional and anatomic definition of cardiac progenitor types. Furthermore, it revealed a cardiac progenitor pool that is anatomically and transcriptionally distinct from currently known cardiac progenitors. Besides contributing to cardiomyocytes, these cells also represent the earliest progenitor of the epicardium, a source of trophic factors and cells during cardiac development and injury. This study provides detailed insights into the formation of early cardiac cell types, with particular relevance to the development of cell-based cardiac regenerative therapies.
Collapse
Affiliation(s)
- Richard C V Tyser
- Department of Physiology, Anatomy and Genetics, South Parks Road, University of Oxford, Oxford OX1 3QX, UK
| | - Ximena Ibarra-Soria
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - Katie McDole
- Howard Hughes Medical Institute, Janelia Research Campus, 19700 Helix Drive, Ashburn, VA 20147, USA
| | - Satish Arcot Jayaram
- Department of Physiology, Anatomy and Genetics, South Parks Road, University of Oxford, Oxford OX1 3QX, UK
| | - Jonathan Godwin
- Department of Physiology, Anatomy and Genetics, South Parks Road, University of Oxford, Oxford OX1 3QX, UK
| | - Teun A H van den Brand
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge CB10 1SD, UK
| | - Antonio M A Miranda
- Department of Physiology, Anatomy and Genetics, South Parks Road, University of Oxford, Oxford OX1 3QX, UK
| | - Antonio Scialdone
- Institute of Epigenetics and Stem Cells, Helmholtz Zentrum München, D-81377 München, Germany
- Institute of Functional Epigenetics, Helmholtz Zentrum München, D-85764 Neuherberg, Germany
- Institute of Computational Biology, Helmholtz Zentrum München, D-85764 Neuherberg, Germany
| | - Philipp J Keller
- Howard Hughes Medical Institute, Janelia Research Campus, 19700 Helix Drive, Ashburn, VA 20147, USA
| | - John C Marioni
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK.
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge CB10 1SD, UK
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK
| | - Shankar Srinivas
- Department of Physiology, Anatomy and Genetics, South Parks Road, University of Oxford, Oxford OX1 3QX, UK.
| |
Collapse
|
23
|
Yoshimi K, Yamauchi Y, Tanaka T, Shimada T, Sato M, Mashimo T. Photoactivatable Cre knock-in mice for spatiotemporal control of genetic engineering in vivo. J Transl Med 2021; 101:125-135. [PMID: 32892213 DOI: 10.1038/s41374-020-00482-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 08/10/2020] [Accepted: 08/11/2020] [Indexed: 11/09/2022] Open
Abstract
Although the Cre-loxP recombination system has been extensively used to analyze gene function in vivo, spatiotemporal control of Cre activity is a critical limitation for easy and precise recombination. Here, we established photoactivatable-Cre (PA-Cre) knock-in (KI) mice at a safe harbor locus for the spatial and temporal regulation of Cre recombinase activity. The mice showed whole-body Cre recombination activity following light exposure for only 1 h. Almost no leaks of Cre recombination activity were detected in the KI mice under natural light conditions. Spot irradiation could induce locus-specific recombination noninvasively, enabling us to compare phenotypes on the left and right sides in the same mouse. Furthermore, long-term irradiation using an implanted wireless LED substantially improved Cre recombination activity, especially in the brain. These results demonstrate that PA-Cre KI mice can facilitate the spatiotemporal control of genetic engineering and provide a useful resource to elucidate gene function in vivo with Cre-loxP.
Collapse
Affiliation(s)
- Kazuto Yoshimi
- Laboratory Animal Research Center, Division of Animal Genetics, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan
- Center for Experimental Medicine and Systems Biology, Division of Genome Engineering, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Yuko Yamauchi
- Laboratory Animal Research Center, Division of Animal Genetics, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan
| | | | | | - Moritoshi Sato
- Graduate School of Arts and Sciences, The University of Tokyo, Tokyo, 153-8902, Japan
| | - Tomoji Mashimo
- Laboratory Animal Research Center, Division of Animal Genetics, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan.
- Center for Experimental Medicine and Systems Biology, Division of Genome Engineering, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan.
- Institute of Experimental Animal Sciences, Graduate School of Medicine, Osaka University, Osaka, 565-0871, Japan.
| |
Collapse
|
24
|
mTOR plays a pivotal role in multiple processes of enamel organ development principally through the mTORC1 pathway and in part via regulating cytoskeleton dynamics. Dev Biol 2020; 467:77-87. [DOI: 10.1016/j.ydbio.2020.08.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 08/04/2020] [Accepted: 08/26/2020] [Indexed: 01/11/2023]
|
25
|
Gheller BJ, Blum JE, Fong EHH, Malysheva OV, Cosgrove BD, Thalacker-Mercer AE. A defined N6-methyladenosine (m 6A) profile conferred by METTL3 regulates muscle stem cell/myoblast state transitions. Cell Death Discov 2020; 6:95. [PMID: 33083017 PMCID: PMC7524727 DOI: 10.1038/s41420-020-00328-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 08/01/2020] [Accepted: 09/02/2020] [Indexed: 01/20/2023] Open
Abstract
Muscle-specific adult stem cells (MuSCs) are required for skeletal muscle regeneration. To ensure efficient skeletal muscle regeneration after injury, MuSCs must undergo state transitions as they are activated from quiescence, give rise to a population of proliferating myoblasts, and continue either to terminal differentiation, to repair or replace damaged myofibers, or self-renewal to repopulate the quiescent population. Changes in MuSC/myoblast state are accompanied by dramatic shifts in their transcriptional profile. Previous reports in other adult stem cell systems have identified alterations in the most abundant internal mRNA modification, N6-methyladenosine (m6A), conferred by its active writer, METTL3, to regulate cell state transitions through alterations in the transcriptional profile of these cells. Our objective was to determine if m6A-modification deposition via METTL3 is a regulator of MuSC/myoblast state transitions in vitro and in vivo. Using liquid chromatography/mass spectrometry we identified that global m6A levels increase during the early stages of skeletal muscle regeneration, in vivo, and decline when C2C12 myoblasts transition from proliferation to differentiation, in vitro. Using m6A-specific RNA-sequencing (MeRIP-seq), a distinct profile of m6A-modification was identified, distinguishing proliferating from differentiating C2C12 myoblasts. RNAi studies show that reducing levels of METTL3, the active m6A methyltransferase, reduced global m6A levels and forced C2C12 myoblasts to prematurely differentiate. Reducing levels of METTL3 in primary mouse MuSCs prior to transplantation enhanced their engraftment capacity upon primary transplantation, however their capacity for serial transplantation was lost. In conclusion, METTL3 regulates m6A levels in MuSCs/myoblasts and controls the transition of MuSCs/myoblasts to different cell states. Furthermore, the first transcriptome wide map of m6A-modifications in proliferating and differentiating C2C12 myoblasts is provided and reveals a number of genes that may regulate MuSC/myoblast state transitions which had not been previously identified.
Collapse
Affiliation(s)
| | - Jamie E. Blum
- Division of Nutritional Sciences, Cornell University, Ithaca, NY USA
| | | | - Olga V. Malysheva
- Division of Nutritional Sciences, Cornell University, Ithaca, NY USA
| | | | - Anna E. Thalacker-Mercer
- Division of Nutritional Sciences, Cornell University, Ithaca, NY USA
- Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, AL USA
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL USA
| |
Collapse
|
26
|
Symmetric neural progenitor divisions require chromatin-mediated homologous recombination DNA repair by Ino80. Nat Commun 2020; 11:3839. [PMID: 32737294 PMCID: PMC7395731 DOI: 10.1038/s41467-020-17551-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 07/02/2020] [Indexed: 12/12/2022] Open
Abstract
Chromatin regulates spatiotemporal gene expression during neurodevelopment, but it also mediates DNA damage repair essential to proliferating neural progenitor cells (NPCs). Here, we uncover molecularly dissociable roles for nucleosome remodeler Ino80 in chromatin-mediated transcriptional regulation and genome maintenance in corticogenesis. We find that conditional Ino80 deletion from cortical NPCs impairs DNA double-strand break (DSB) repair, triggering p53-dependent apoptosis and microcephaly. Using an in vivo DSB repair pathway assay, we find that Ino80 is selectively required for homologous recombination (HR) DNA repair, which is mechanistically distinct from Ino80 function in YY1-associated transcription. Unexpectedly, sensitivity to loss of Ino80-mediated HR is dependent on NPC division mode: Ino80 deletion leads to unrepaired DNA breaks and apoptosis in symmetric NPC-NPC divisions, but not in asymmetric neurogenic divisions. This division mode dependence is phenocopied following conditional deletion of HR gene Brca2. Thus, distinct modes of NPC division have divergent requirements for Ino80-dependent HR DNA repair.
Collapse
|
27
|
Martin RA, Buckley KH, Mankowski DC, Riley BM, Sidwell AN, Douglas SL, Worth RG, Pizza FX. Myogenic Cell Expression of Intercellular Adhesion Molecule-1 Contributes to Muscle Regeneration after Injury. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:2039-2055. [PMID: 32650005 DOI: 10.1016/j.ajpath.2020.06.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 06/10/2020] [Accepted: 06/15/2020] [Indexed: 10/23/2022]
Abstract
This study investigated intercellular adhesion molecule-1 (ICAM-1), a membrane protein that mediates cell-to-cell adhesion and communication, as a mechanism through which the inflammatory response facilitates muscle regeneration after injury. Toxin-induced muscle injury to tibialis anterior muscles of wild-type mice caused ICAM-1 to be expressed by a population of satellite cells/myoblasts and myofibers. Myogenic cell expression of ICAM-1 contributed to the restoration of muscle structure after injury, as regenerating myofibers were more abundant and myofiber size was larger for wild-type compared with Icam1-/- mice during 28 days of recovery. Contrastingly, restoration of muscle function after injury was similar between the genotypes. ICAM-1 facilitated the restoration of muscle structure after injury through mechanisms involving the regulation of myofiber branching, protein synthesis, and the organization of nuclei within myofibers after myogenic cell fusion. These findings provide support for a paradigm in which ICAM-1 expressed by myogenic cells after muscle injury augments their adhesive and fusogenic properties, which, in turn, facilitates regenerative and hypertrophic processes that restore structure to injured muscle.
Collapse
Affiliation(s)
- Ryan A Martin
- School of Exercise and Rehabilitation Sciences, The University of Toledo, Toledo, Ohio
| | - Kole H Buckley
- School of Exercise and Rehabilitation Sciences, The University of Toledo, Toledo, Ohio
| | - Drew C Mankowski
- School of Exercise and Rehabilitation Sciences, The University of Toledo, Toledo, Ohio
| | - Benjamin M Riley
- School of Exercise and Rehabilitation Sciences, The University of Toledo, Toledo, Ohio
| | - Alena N Sidwell
- School of Exercise and Rehabilitation Sciences, The University of Toledo, Toledo, Ohio
| | - Stephanie L Douglas
- School of Exercise and Rehabilitation Sciences, The University of Toledo, Toledo, Ohio
| | - Randall G Worth
- Department of Medical Microbiology and Immunology, The University of Toledo, Toledo, Ohio
| | - Francis X Pizza
- School of Exercise and Rehabilitation Sciences, The University of Toledo, Toledo, Ohio.
| |
Collapse
|
28
|
Rueda EM, Hall BM, Hill MC, Swinton PG, Tong X, Martin JF, Poché RA. The Hippo Pathway Blocks Mammalian Retinal Müller Glial Cell Reprogramming. Cell Rep 2020; 27:1637-1649.e6. [PMID: 31067451 PMCID: PMC6521882 DOI: 10.1016/j.celrep.2019.04.047] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 02/04/2019] [Accepted: 04/09/2019] [Indexed: 02/08/2023] Open
Abstract
In response to retinal damage, the Müller glial cells (MGs) of the zebrafish retina have the ability to undergo a cellular reprogramming event in which they enter the cell cycle and divide asymmetrically, thereby producing multipotent retinal progenitors capable of regenerating lost retinal neurons. However, mammalian MGs do not exhibit such a proliferative and regenerative ability. Here, we identify Hippo pathway-mediated repression of the transcription cofactor YAP as a core regulatory mechanism that normally blocks mammalian MG proliferation and cellular reprogramming. MG-specific deletion of Hippo pathway components Lats1 and Lats2, as well as transgenic expression of a Hippo non-responsive form of YAP (YAP5SA), resulted in dramatic Cyclin D1 upregulation, loss of adult MG identity, and attainment of a highly proliferative, progenitor-like cellular state. Our results reveal that mammalian MGs may have latent regenerative capacity that can be stimulated by repressing Hippo signaling. Rueda et al. identify the Hippo pathway as an endogenous molecular mechanism normally preventing mammalian Müller glial reprogramming to a proliferative, progenitor-like state.
Collapse
Affiliation(s)
- Elda M Rueda
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Benjamin M Hall
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA; Graduate Program in Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Matthew C Hill
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA; Graduate Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Paul G Swinton
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA; Texas Heart Institute, Cardiomyocyte Renewal Lab, Houston, TX 77030, USA
| | - Xuefei Tong
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - James F Martin
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA; Graduate Program in Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA; Graduate Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA; Development, Disease Models and Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA; Genetics and Genomics Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA; Cardiovasular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA; Texas Heart Institute, Cardiomyocyte Renewal Lab, Houston, TX 77030, USA.
| | - Ross A Poché
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA; Graduate Program in Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA; Graduate Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA; Development, Disease Models and Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA; Genetics and Genomics Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
29
|
Ikonomou L, Herriges MJ, Lewandowski SL, Marsland R, Villacorta-Martin C, Caballero IS, Frank DB, Sanghrajka RM, Dame K, Kańduła MM, Hicks-Berthet J, Lawton ML, Christodoulou C, Fabian AJ, Kolaczyk E, Varelas X, Morrisey EE, Shannon JM, Mehta P, Kotton DN. The in vivo genetic program of murine primordial lung epithelial progenitors. Nat Commun 2020; 11:635. [PMID: 32005814 PMCID: PMC6994558 DOI: 10.1038/s41467-020-14348-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 12/23/2019] [Indexed: 12/29/2022] Open
Abstract
Multipotent Nkx2-1-positive lung epithelial primordial progenitors of the foregut endoderm are thought to be the developmental precursors to all adult lung epithelial lineages. However, little is known about the global transcriptomic programs or gene networks that regulate these gateway progenitors in vivo. Here we use bulk RNA-sequencing to describe the unique genetic program of in vivo murine lung primordial progenitors and computationally identify signaling pathways, such as Wnt and Tgf-β superfamily pathways, that are involved in their cell-fate determination from pre-specified embryonic foregut. We integrate this information in computational models to generate in vitro engineered lung primordial progenitors from mouse pluripotent stem cells, improving the fidelity of the resulting cells through unbiased, easy-to-interpret similarity scores and modulation of cell culture conditions, including substratum elastic modulus and extracellular matrix composition. The methodology proposed here can have wide applicability to the in vitro derivation of bona fide tissue progenitors of all germ layers.
Collapse
Affiliation(s)
- Laertis Ikonomou
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA, 02118, USA.
- The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA, 02118, USA.
| | - Michael J Herriges
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA, 02118, USA
- The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Sara L Lewandowski
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA, 02118, USA
- The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Robert Marsland
- Department of Physics, Boston University, Boston, MA, 02215, USA
| | - Carlos Villacorta-Martin
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA, 02118, USA
| | - Ignacio S Caballero
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA, 02118, USA
| | - David B Frank
- Division of Pediatric Cardiology, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Reeti M Sanghrajka
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA, 02118, USA
- The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Keri Dame
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA, 02118, USA
- The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Maciej M Kańduła
- Department of Mathematics & Statistics, Boston University, Boston, MA, 02215, USA
- Chair of Bioinformatics Research Group, Boku University, 1190, Vienna, Austria
| | - Julia Hicks-Berthet
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Matthew L Lawton
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA, 02118, USA
- The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Constantina Christodoulou
- The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA, 02118, USA
| | | | - Eric Kolaczyk
- Department of Mathematics & Statistics, Boston University, Boston, MA, 02215, USA
| | - Xaralabos Varelas
- Department of Biochemistry, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Edward E Morrisey
- Penn Center for Pulmonary Biology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - John M Shannon
- Division of Pulmonary Biology, Cincinnati Children's Hospital, Cincinnati, OH, 45229, USA
| | - Pankaj Mehta
- Department of Physics, Boston University, Boston, MA, 02215, USA
| | - Darrell N Kotton
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA, 02118, USA.
- The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA, 02118, USA.
| |
Collapse
|
30
|
Wang J, Batourina E, Schneider K, Souza S, Swayne T, Liu C, George CD, Tate T, Dan H, Wiessner G, Zhuravlev Y, Canman JC, Mysorekar IU, Mendelsohn CL. Polyploid Superficial Cells that Maintain the Urothelial Barrier Are Produced via Incomplete Cytokinesis and Endoreplication. Cell Rep 2019; 25:464-477.e4. [PMID: 30304685 PMCID: PMC6351079 DOI: 10.1016/j.celrep.2018.09.042] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 05/28/2018] [Accepted: 09/12/2018] [Indexed: 01/26/2023] Open
Abstract
The urothelium is an epithelia barrier lined by a luminal layer of binucleated, octoploid, superficial cells. Superficial cells are critical for production and transport of uroplakins, a family of proteins that assemble into a waterproof crystalline plaque that helps protect against infection and toxic substances. Adult urothelium is nearly quiescent, but rapidly regenerates in response to injury. Yet the mechanism by which binucleated, polyploid, superficial cells are produced remains unclear. Here, we show that superficial cells are likely to be derived from a population of binucleated intermediate cells, which are produced from mononucleated intermediate cells via incomplete cytokinesis. We show that binucleated intermediate and superficial cells increase DNA content via endoreplication, passing through S phase without entering mitosis. The urothelium can be permanently damaged by repetitive or chronic injury or disease. Identification of the mechanism by which superficial cells are produced may be important for developing strategies for urothelial repair.
Collapse
Affiliation(s)
- Jia Wang
- Department of Urology, Columbia University, 1130 St. Nicholas Avenue, New York, NY 10032, USA
| | - Ekatherina Batourina
- Department of Urology, Genetics, and Development and Pathology and Cell Biology, Columbia University, 1130 St. Nicholas Avenue, New York, NY 10032, USA
| | - Kerry Schneider
- College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Spenser Souza
- Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA 70112, USA
| | - Theresa Swayne
- Confocal and Specialized Microscopy Shared Resource, Herbert Irving Comprehensive Cancer Center, Columbia University, 1130 St. Nicholas Avenue, New York, NY 10032, USA
| | - Chang Liu
- Department of Urology, Genetics, and Development and Pathology and Cell Biology, Columbia University, 1130 St. Nicholas Avenue, New York, NY 10032, USA
| | - Christopher D George
- Department of Urology, Genetics, and Development and Pathology and Cell Biology, Columbia University, 1130 St. Nicholas Avenue, New York, NY 10032, USA
| | - Tiffany Tate
- Department of Urology, Genetics, and Development and Pathology and Cell Biology, Columbia University, 1130 St. Nicholas Avenue, New York, NY 10032, USA
| | - Hanbin Dan
- Department of Systems Biology, Columbia University, New York, NY 10032, USA
| | - Gregory Wiessner
- Department of Urology, Genetics, and Development and Pathology and Cell Biology, Columbia University, 1130 St. Nicholas Avenue, New York, NY 10032, USA
| | - Yelena Zhuravlev
- Department of Urology, Genetics, and Development and Pathology and Cell Biology, Columbia University, 1130 St. Nicholas Avenue, New York, NY 10032, USA
| | - Julie C Canman
- Department of Pathology and Cell Biology, Columbia University, 630 West 168th Street, New York, NY 10032, USA
| | - Indira U Mysorekar
- Departments of Obstetrics and Gynecology and Pathology and Immunology and Center for Reproductive Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Cathy Lee Mendelsohn
- Department of Urology, Genetics, and Development and Pathology and Cell Biology, Columbia University, 1130 St. Nicholas Avenue, New York, NY 10032, USA.
| |
Collapse
|
31
|
Sakai M, Troutman TD, Seidman JS, Ouyang Z, Spann NJ, Abe Y, Ego KM, Bruni CM, Deng Z, Schlachetzki JCM, Nott A, Bennett H, Chang J, Vu BT, Pasillas MP, Link VM, Texari L, Heinz S, Thompson BM, McDonald JG, Geissmann F, Glass CK. Liver-Derived Signals Sequentially Reprogram Myeloid Enhancers to Initiate and Maintain Kupffer Cell Identity. Immunity 2019; 51:655-670.e8. [PMID: 31587991 DOI: 10.1016/j.immuni.2019.09.002] [Citation(s) in RCA: 253] [Impact Index Per Article: 42.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 07/27/2019] [Accepted: 09/03/2019] [Indexed: 12/20/2022]
Abstract
Tissue environment plays a powerful role in establishing and maintaining the distinct phenotypes of resident macrophages, but the underlying molecular mechanisms remain poorly understood. Here, we characterized transcriptomic and epigenetic changes in repopulating liver macrophages following acute Kupffer cell depletion as a means to infer signaling pathways and transcription factors that promote Kupffer cell differentiation. We obtained evidence that combinatorial interactions of the Notch ligand DLL4 and transforming growth factor-b (TGF-β) family ligands produced by sinusoidal endothelial cells and endogenous LXR ligands were required for the induction and maintenance of Kupffer cell identity. DLL4 regulation of the Notch transcriptional effector RBPJ activated poised enhancers to rapidly induce LXRα and other Kupffer cell lineage-determining factors. These factors in turn reprogrammed the repopulating liver macrophage enhancer landscape to converge on that of the original resident Kupffer cells. Collectively, these findings provide a framework for understanding how macrophage progenitor cells acquire tissue-specific phenotypes.
Collapse
Affiliation(s)
- Mashito Sakai
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Ty D Troutman
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jason S Seidman
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Zhengyu Ouyang
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Nathanael J Spann
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Yohei Abe
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Kaori M Ego
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Cassi M Bruni
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Zihou Deng
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Johannes C M Schlachetzki
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Alexi Nott
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Hunter Bennett
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jonathan Chang
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - BaoChau T Vu
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Martina P Pasillas
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Verena M Link
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Faculty of Biology, Division of Evolutionary Biology, Ludwig-Maximilian University of Munich, Munich 82152, Germany
| | - Lorane Texari
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Sven Heinz
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Bonne M Thompson
- Department of Molecular Genetics, Center for Human Nutrition, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jeffrey G McDonald
- Department of Molecular Genetics, Center for Human Nutrition, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Frederic Geissmann
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Christopher K Glass
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
32
|
Lopez AL, Larina IV. Second harmonic generation microscopy of early embryonic mouse hearts. BIOMEDICAL OPTICS EXPRESS 2019; 10:2898-2908. [PMID: 31259060 PMCID: PMC6583332 DOI: 10.1364/boe.10.002898] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 05/15/2019] [Accepted: 05/16/2019] [Indexed: 05/15/2023]
Abstract
The understanding of biomechanical regulation of early heart development in genetic mouse models can contribute to improved management of congenital cardiovascular defects and embryonic cardiac failures in humans. The extracellular matrix (ECM), and particularly fibrillar collagen, are central to heart biomechanics, regulating tissue strength, elasticity and contractility. Here, we explore second harmonic generation (SHG) microscopy for visualization of establishing cardiac fibers such as collagen in mouse embryos through the earliest stages of development. We detected significant increase in SHG positive fibrillar content and organization over the first 24 hours after initiation of contractions. SHG microscopy revealed regions of higher fibrillar organization in regions of higher contractility and reduced fibrillar content and organization in mouse Mlc2a model with cardiac contractility defect, suggesting regulatory role of mechanical load in production and organization of structural fibers from the earliest stages. Simultaneous volumetric SHG and two-photon excitation microscopy of vital fluorescent reporter EGFP in the heart was demonstrated. In summary, these data set SHG microscopy as a valuable non-bias imaging tool to investigate mouse embryonic cardiogenesis and biomechanics.
Collapse
Affiliation(s)
- Andrew L. Lopez
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Irina V. Larina
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| |
Collapse
|
33
|
Abstract
Cell-type-specific gene targeting with the Cre/loxP system has become an indispensable technique in experimental neuroscience, particularly for the study of late-born glial cells that make myelin. A plethora of conditional mutants and Cre-expressing mouse lines is now available to the research community that allows laboratories to readily engage in in vivo analyses of oligodendrocytes and their precursor cells. This chapter summarizes concepts and strategies in targeting myelinating glial cells in mice for mutagenesis or imaging, and provides an overview of the most important Cre driver lines successfully used in this rapidly growing field.
Collapse
Affiliation(s)
- Sandra Goebbels
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany.
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany.
| |
Collapse
|
34
|
Wang YX, Feige P, Brun CE, Hekmatnejad B, Dumont NA, Renaud JM, Faulkes S, Guindon DE, Rudnicki MA. EGFR-Aurka Signaling Rescues Polarity and Regeneration Defects in Dystrophin-Deficient Muscle Stem Cells by Increasing Asymmetric Divisions. Cell Stem Cell 2019; 24:419-432.e6. [PMID: 30713094 DOI: 10.1016/j.stem.2019.01.002] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 11/20/2018] [Accepted: 01/07/2019] [Indexed: 12/17/2022]
Abstract
Loss of dystrophin expression in Duchenne muscular dystrophy (DMD) causes progressive degeneration of skeletal muscle, which is exacerbated by reduced self-renewing asymmetric divisions of muscle satellite cells. This, in turn, affects the production of myogenic precursors and impairs regeneration and suggests that increasing such divisions may be beneficial. Here, through a small-molecule screen, we identified epidermal growth factor receptor (EGFR) and Aurora kinase A (Aurka) as regulators of asymmetric satellite cell divisions. Inhibiting EGFR causes a substantial shift from asymmetric to symmetric division modes, whereas EGF treatment increases asymmetric divisions. EGFR activation acts through Aurka to orient mitotic centrosomes, and inhibiting Aurka blocks EGF stimulation-induced asymmetric division. In vivo EGF treatment markedly activates asymmetric divisions of dystrophin-deficient satellite cells in mdx mice, increasing progenitor numbers, enhancing regeneration, and restoring muscle strength. Therefore, activating an EGFR-dependent polarity pathway promotes functional rescue of dystrophin-deficient satellite cells and enhances muscle force generation.
Collapse
Affiliation(s)
- Yu Xin Wang
- Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, ON, Canada; Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Peter Feige
- Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, ON, Canada; Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Caroline E Brun
- Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, ON, Canada; Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Bahareh Hekmatnejad
- Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, ON, Canada; Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Nicolas A Dumont
- Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, ON, Canada; Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Jean-Marc Renaud
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Sharlene Faulkes
- Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, ON, Canada; Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Daniel E Guindon
- Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, ON, Canada; Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Michael A Rudnicki
- Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, ON, Canada; Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Department of Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
35
|
Lukjanenko L, Karaz S, Stuelsatz P, Gurriaran-Rodriguez U, Michaud J, Dammone G, Sizzano F, Mashinchian O, Ancel S, Migliavacca E, Liot S, Jacot G, Metairon S, Raymond F, Descombes P, Palini A, Chazaud B, Rudnicki MA, Bentzinger CF, Feige JN. Aging Disrupts Muscle Stem Cell Function by Impairing Matricellular WISP1 Secretion from Fibro-Adipogenic Progenitors. Cell Stem Cell 2019; 24:433-446.e7. [PMID: 30686765 DOI: 10.1016/j.stem.2018.12.014] [Citation(s) in RCA: 203] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 10/15/2018] [Accepted: 12/17/2018] [Indexed: 12/17/2022]
Abstract
Research on age-related regenerative failure of skeletal muscle has extensively focused on the phenotypes of muscle stem cells (MuSCs). In contrast, the impact of aging on regulatory cells in the MuSC niche remains largely unexplored. Here, we demonstrate that aging impairs the function of mouse fibro-adipogenic progenitors (FAPs) and thereby indirectly affects the myogenic potential of MuSCs. Using transcriptomic profiling, we identify WNT1 Inducible Signaling Pathway Protein 1 (WISP1) as a FAP-derived matricellular signal that is lost during aging. WISP1 is required for efficient muscle regeneration and controls the expansion and asymmetric commitment of MuSCs through Akt signaling. Transplantation of young FAPs or systemic treatment with WISP1 restores the myogenic capacity of MuSCs in aged mice and rescues skeletal muscle regeneration. Our work establishes that loss of WISP1 from FAPs contributes to MuSC dysfunction in aged skeletal muscles and demonstrates that this mechanism can be targeted to rejuvenate myogenesis.
Collapse
Affiliation(s)
- Laura Lukjanenko
- Nestlé Research, EPFL Innovation Park, 1015 Lausanne, Switzerland; School of Life Sciences, Ecole Polytechnique Federale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Sonia Karaz
- Nestlé Research, EPFL Innovation Park, 1015 Lausanne, Switzerland
| | - Pascal Stuelsatz
- Nestlé Research, EPFL Innovation Park, 1015 Lausanne, Switzerland
| | - Uxia Gurriaran-Rodriguez
- Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, ON K1H8L6, Canada; Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Joris Michaud
- Nestlé Research, EPFL Innovation Park, 1015 Lausanne, Switzerland
| | - Gabriele Dammone
- Nestlé Research, EPFL Innovation Park, 1015 Lausanne, Switzerland
| | - Federico Sizzano
- Nestlé Research, EPFL Innovation Park, 1015 Lausanne, Switzerland
| | - Omid Mashinchian
- Nestlé Research, EPFL Innovation Park, 1015 Lausanne, Switzerland; School of Life Sciences, Ecole Polytechnique Federale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Sara Ancel
- Nestlé Research, EPFL Innovation Park, 1015 Lausanne, Switzerland; School of Life Sciences, Ecole Polytechnique Federale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | | | - Sophie Liot
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, CNRS 5310, INSERM U1217, Lyon, France
| | - Guillaume Jacot
- Nestlé Research, EPFL Innovation Park, 1015 Lausanne, Switzerland
| | | | - Frederic Raymond
- Nestlé Research, EPFL Innovation Park, 1015 Lausanne, Switzerland
| | | | - Alessio Palini
- Nestlé Research, EPFL Innovation Park, 1015 Lausanne, Switzerland
| | - Benedicte Chazaud
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, CNRS 5310, INSERM U1217, Lyon, France
| | - Michael A Rudnicki
- Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, ON K1H8L6, Canada; Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - C Florian Bentzinger
- Nestlé Research, EPFL Innovation Park, 1015 Lausanne, Switzerland; Département de pharmacologie et physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Jerome N Feige
- Nestlé Research, EPFL Innovation Park, 1015 Lausanne, Switzerland; School of Life Sciences, Ecole Polytechnique Federale de Lausanne (EPFL), 1015 Lausanne, Switzerland.
| |
Collapse
|
36
|
Chang CN, Singh AJ, Gross MK, Kioussi C. Requirement of Pitx2 for skeletal muscle homeostasis. Dev Biol 2019; 445:90-102. [PMID: 30414844 PMCID: PMC6289786 DOI: 10.1016/j.ydbio.2018.11.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 11/05/2018] [Accepted: 11/05/2018] [Indexed: 12/24/2022]
Abstract
Skeletal muscle is generated by the successive incorporation of primary (embryonic), secondary (fetal), and tertiary (adult) fibers into muscle. Conditional excision of Pitx2 function by an MCKCre driver resulted in animals with histological and ultrastructural defects in P30 muscles and fibers, respectively. Mutant muscle showed severe reduction in mitochondria and FoxO3-mediated mitophagy. Both oxidative and glycolytic energy metabolism were reduced. Conditional excision was limited to fetal muscle fibers after the G1-G0 transition and resulted in altered MHC, Rac1, MEF2a, and alpha-tubulin expression within these fibers. The onset of excision, monitored by a nuclear reporter gene, was observed as early as E16. Muscle at this stage was already severely malformed, but appeared to recover by P30 by the expansion of adjoining larger fibers. Our studies demonstrate that the homeodomain transcription factor Pitx2 has a postmitotic role in maintaining skeletal muscle integrity and energy homeostasis in fetal muscle fibers.
Collapse
Affiliation(s)
- Chih-Ning Chang
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA; Molecular Cell Biology Graduate Program, Oregon State University, Corvallis, OR 97331, USA
| | - Arun J Singh
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA
| | - Michael K Gross
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA
| | - Chrissa Kioussi
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA.
| |
Collapse
|
37
|
Bachman JF, Klose A, Liu W, Paris ND, Blanc RS, Schmalz M, Knapp E, Chakkalakal JV. Prepubertal skeletal muscle growth requires Pax7-expressing satellite cell-derived myonuclear contribution. Development 2018; 145:dev.167197. [PMID: 30305290 PMCID: PMC6215399 DOI: 10.1242/dev.167197] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 09/18/2018] [Indexed: 12/14/2022]
Abstract
The functional role of Pax7-expressing satellite cells (SCs) in postnatal skeletal muscle development beyond weaning remains obscure. Therefore, the relevance of SCs during prepubertal growth, a period after weaning but prior to the onset of puberty, has not been examined. Here, we have characterized mouse skeletal muscle growth during prepuberty and found significant increases in myofiber cross-sectional area that correlated with SC-derived myonuclear number. Remarkably, genome-wide RNA-sequencing analysis established that post-weaning juvenile and early adolescent skeletal muscle have markedly different gene expression signatures. These distinctions are consistent with extensive skeletal muscle maturation during this essential, albeit brief, developmental phase. Indelible labeling of SCs with Pax7CreERT2/+; Rosa26nTnG/+ mice demonstrated SC-derived myonuclear contribution during prepuberty, with a substantial reduction at puberty onset. Prepubertal depletion of SCs in Pax7CreERT2/+; Rosa26DTA/+ mice reduced myofiber size and myonuclear number, and caused force generation deficits to a similar extent in both fast and slow-contracting muscles. Collectively, these data demonstrate SC-derived myonuclear accretion as a cellular mechanism that contributes to prepubertal hypertrophic skeletal muscle growth. Summary: Examination of gene expression and morphological changes in mouse skeletal muscle during prepuberty demonstrates that satellite cell-derived myonuclear accretion contributes to prepubertal hypertrophic skeletal muscle growth.
Collapse
Affiliation(s)
- John F Bachman
- Department of Pathology and Laboratory Medicine, Cell Biology of Disease Graduate Program, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY 14642, USA.,Department of Pharmacology and Physiology, University of Rochester Medical Center, 601 Elmwood Ave Box 711, Rochester, NY 14642, USA
| | - Alanna Klose
- Department of Pharmacology and Physiology, University of Rochester Medical Center, 601 Elmwood Ave Box 711, Rochester, NY 14642, USA
| | - Wenxuan Liu
- Department of Pharmacology and Physiology, University of Rochester Medical Center, 601 Elmwood Ave Box 711, Rochester, NY 14642, USA .,Department of Biomedical Genetics, Genetics, Development, and Stem Cells Graduate Program, University of Rochester Medical Center, 601 Elmwood Ave Box 633, Rochester, NY 14642, USA
| | - Nicole D Paris
- Department of Pharmacology and Physiology, University of Rochester Medical Center, 601 Elmwood Ave Box 711, Rochester, NY 14642, USA
| | - Roméo S Blanc
- Department of Pharmacology and Physiology, University of Rochester Medical Center, 601 Elmwood Ave Box 711, Rochester, NY 14642, USA
| | - Melissa Schmalz
- Department of Pharmacology and Physiology, University of Rochester Medical Center, 601 Elmwood Ave Box 711, Rochester, NY 14642, USA
| | - Emma Knapp
- Department of Orthopaedics and Rehabilitation, Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY 14642, USA
| | - Joe V Chakkalakal
- Department of Pharmacology and Physiology, University of Rochester Medical Center, 601 Elmwood Ave Box 711, Rochester, NY 14642, USA.,Wilmot Cancer Institute, Stem Cell and Regenerative Medicine Institute, The Rochester Aging Research Center, and Department of Biomedical Engineering, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY 14642, USA
| |
Collapse
|
38
|
Nie X, Zheng J, Ricupero CL, He L, Jiao K, Mao JJ. mTOR acts as a pivotal signaling hub for neural crest cells during craniofacial development. PLoS Genet 2018; 14:e1007491. [PMID: 29975682 PMCID: PMC6049956 DOI: 10.1371/journal.pgen.1007491] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 07/17/2018] [Accepted: 06/14/2018] [Indexed: 12/14/2022] Open
Abstract
mTOR is a highly conserved serine/threonine protein kinase that is critical for diverse cellular processes in both developmental and physiological settings. mTOR interacts with a set of molecules including Raptor and Rictor to form two distinct functional complexes, namely the mTORC1 and mTORC2. Here, we used novel genetic models to investigate functions of the mTOR pathway for cranial neural crest cells (NCCs), which are a temporary type of cells arising from the ectoderm layer and migrate to the pharyngeal arches participating craniofacial development. mTOR deletion elicited a proliferation deficit and excessive apoptosis of post-migratory NCCs, leading to growth arrest of the facial primordia along with midline orofacial clefts. Furthermore, NCC differentiation was impaired. Thus, NCC derivatives, such as skeletons, vasculatures and neural tissues were either rudimentary or malformed. We further demonstrate that disruption of mTOR caused P53 hyperactivity and cell cycle arrest in cranial NCCs, and lowering P53 activity by one copy reduction attenuated the severity of craniofacial phenotype in NCC-mTOR knockout mice. Remarkably, NCC-Rptor disruption caused a spectrum of defects mirroring that of the NCC-mTOR deletion, whereas NCC-Rictor disruption only caused a mild craniofacial phenotype compared to the mTOR and Rptor conditional knockout models. Altogether, our data demonstrate that mTOR functions mediated by mTORC1 are indispensable for multiple processes of NCC development including proliferation, survival, and differentiation during craniofacial morphogenesis and organogenesis, and P53 hyperactivity in part accounts for the defective craniofacial development in NCC-mTOR knockout mice.
Collapse
Affiliation(s)
- Xuguang Nie
- Center for Craniofacial Regeneration, College of Dental Medicine, Columbia University, New York, New York, United States of America
| | - Jinxuan Zheng
- Center for Craniofacial Regeneration, College of Dental Medicine, Columbia University, New York, New York, United States of America
| | - Christopher L. Ricupero
- Center for Craniofacial Regeneration, College of Dental Medicine, Columbia University, New York, New York, United States of America
| | - Ling He
- Center for Craniofacial Regeneration, College of Dental Medicine, Columbia University, New York, New York, United States of America
| | - Kai Jiao
- University of Alabama at Birmingham, Department of Genetics, Birmingham, Alabama, United States of America
| | - Jeremy J. Mao
- Center for Craniofacial Regeneration, College of Dental Medicine, Columbia University, New York, New York, United States of America
- Department of Pathology and Cell Biology, Columbia University, New York, New York, United States of America
| |
Collapse
|
39
|
Cutler AA, Jackson JB, Corbett AH, Pavlath GK. Non-equivalence of nuclear import among nuclei in multinucleated skeletal muscle cells. J Cell Sci 2018; 131:jcs.207670. [PMID: 29361530 DOI: 10.1242/jcs.207670] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 12/21/2017] [Indexed: 01/01/2023] Open
Abstract
Skeletal muscle is primarily composed of large myofibers containing thousands of post-mitotic nuclei distributed throughout a common cytoplasm. Protein production and localization in specialized myofiber regions is crucial for muscle function. Myonuclei differ in transcriptional activity and protein accumulation, but how these differences among nuclei sharing a cytoplasm are achieved is unknown. Regulated nuclear import of proteins is one potential mechanism for regulating transcription spatially and temporally in individual myonuclei. The best-characterized nuclear localization signal (NLS) in proteins is the classical NLS (cNLS), but many other NLS motifs exist. We examined cNLS and non-cNLS reporter protein import using multinucleated muscle cells generated in vitro, revealing that cNLS and non-cNLS nuclear import differs among nuclei in the same cell. Investigation of cNLS nuclear import rates in isolated myofibers ex vivo confirmed differences in nuclear import rates among myonuclei. Analyzing nuclear import throughout myogenesis revealed that cNLS and non-cNLS import varies during differentiation. Taken together, our results suggest that both spatial and temporal regulation of nuclear import pathways are important in muscle cell differentiation and protein regionalization in myofibers.
Collapse
Affiliation(s)
- Alicia A Cutler
- Department of Pharmacology, Emory University, Atlanta, GA 30322, USA.,Graduate Program in Biochemistry, Cell and Developmental Biology, Emory University, Atlanta, GA 30322, USA
| | | | - Anita H Corbett
- Department of Biology, Emory University, Atlanta, GA 30322, USA
| | - Grace K Pavlath
- Department of Pharmacology, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
40
|
Cates J, Nevell L, Prajapati SI, Nelon LD, Chang JY, Randolph ME, Wood B, Keller C, Whitaker RT. Shape analysis of the basioccipital bone in Pax7-deficient mice. Sci Rep 2017; 7:17955. [PMID: 29263370 PMCID: PMC5738401 DOI: 10.1038/s41598-017-18199-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 12/04/2017] [Indexed: 11/09/2022] Open
Abstract
We compared the cranial base of newborn Pax7-deficient and wildtype mice using a computational shape modeling technology called particle-based modeling (PBM). We found systematic differences in the morphology of the basiooccipital bone, including a broadening of the basioccipital bone and an antero-inferior inflection of its posterior edge in the Pax7-deficient mice. We show that the Pax7 cell lineage contributes to the basioccipital bone and that the location of the Pax7 lineage correlates with the morphology most effected by Pax7 deficiency. Our results suggest that the Pax7-deficient mouse may be a suitable model for investigating the genetic control of the location and orientation of the foramen magnum, and changes in the breadth of the basioccipital.
Collapse
Affiliation(s)
- Joshua Cates
- Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, UT, USA
| | - Lisa Nevell
- Department of Anthropology, Center for the Advanced Study of Human Paleobiology, The George Washington University, Washington DC, USA.
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center, San Antonio, TX, USA.
| | - Suresh I Prajapati
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center, San Antonio, TX, USA
| | - Laura D Nelon
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center, San Antonio, TX, USA
| | - Jerry Y Chang
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center, San Antonio, TX, USA
| | | | - Bernard Wood
- Department of Anthropology, Center for the Advanced Study of Human Paleobiology, The George Washington University, Washington DC, USA
| | - Charles Keller
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center, San Antonio, TX, USA.
- Children's Cancer Therapy Development Institute, Beaverton, OR, USA.
| | - Ross T Whitaker
- Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
41
|
Cutler AA, Dammer EB, Doung DM, Seyfried NT, Corbett AH, Pavlath GK. Biochemical isolation of myonuclei employed to define changes to the myonuclear proteome that occur with aging. Aging Cell 2017; 16:738-749. [PMID: 28544616 PMCID: PMC5506426 DOI: 10.1111/acel.12604] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2017] [Indexed: 01/03/2023] Open
Abstract
Skeletal muscle aging is accompanied by loss of muscle mass and strength. Examining changes in myonuclear proteins with age would provide insight into molecular processes which regulate these profound changes in muscle physiology. However, muscle tissue is highly adapted for contraction and thus comprised largely of contractile proteins making the nuclear proteins difficult to identify from whole muscle samples. By developing a method to purify myonuclei from whole skeletal muscle, we were able to collect myonuclei for analysis by flow cytometry, biochemistry, and mass spectrometry. Nuclear purification dramatically increased the number and intensity of nuclear proteins detected by mass spectrometry compared to whole tissue. We exploited this increased proteomic depth to investigate age-related changes to the myonuclear proteome. Nuclear levels of 54 of 779 identified proteins (7%) changed significantly with age; these proteins were primarily involved in chromatin maintenance and RNA processing. To determine whether the changes we detected were specific to myonuclei or were common to nuclei of excitatory tissues, we compared aging in myonuclei to aging in brain nuclei. Although several of the same processes were affected by aging in both brain and muscle nuclei, the specific proteins involved in these alterations differed between the two tissues. Isolating myonuclei allowed a deeper view into the myonuclear proteome than previously possible facilitating identification of novel age-related changes in skeletal muscle. Our technique will enable future studies into a heretofore underrepresented compartment of skeletal muscle.
Collapse
Affiliation(s)
- Alicia A. Cutler
- Department of Pharmacology; Emory University; Atlanta GA 30322 USA
- Graduate Program in Biochemistry, Cell and Developmental Biology; Emory University; Atlanta GA 30322 USA
| | - Eric B. Dammer
- Department of Biochemistry; Emory University; Atlanta GA 30322 USA
| | - Duc M. Doung
- Department of Biochemistry; Emory University; Atlanta GA 30322 USA
| | | | | | - Grace K. Pavlath
- Department of Pharmacology; Emory University; Atlanta GA 30322 USA
| |
Collapse
|
42
|
Hill RA, Damisah EC, Chen F, Kwan AC, Grutzendler J. Targeted two-photon chemical apoptotic ablation of defined cell types in vivo. Nat Commun 2017. [PMID: 28621306 PMCID: PMC5501159 DOI: 10.1038/ncomms15837] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
A major bottleneck limiting understanding of mechanisms and consequences of cell death in complex organisms is the inability to induce and visualize this process with spatial and temporal precision in living animals. Here we report a technique termed two-photon chemical apoptotic targeted ablation (2Phatal) that uses focal illumination with a femtosecond-pulsed laser to bleach a nucleic acid-binding dye causing dose-dependent apoptosis of individual cells without collateral damage. Using 2Phatal, we achieve precise ablation of distinct populations of neurons, glia and pericytes in the mouse brain and in zebrafish. When combined with organelle-targeted fluorescent proteins and biosensors, we uncover previously unrecognized cell-type differences in patterns of apoptosis and associated dynamics of ribosomal disassembly, calcium overload and mitochondrial fission. 2Phatal provides a powerful and rapidly adoptable platform to investigate in vivo functional consequences and neural plasticity following cell death as well as apoptosis, cell clearance and tissue remodelling in diverse organs and species. Investigating cell death in living organisms is hampered by a lack of techniques to induce apoptosis with spatial and temporal precision without collateral damage. Here the authors develop two-photon chemical apoptotic targeted ablation (2Phatal), allowing studies of apoptosis and its functional consequences in vivo.
Collapse
Affiliation(s)
- Robert A Hill
- Department of Neurology, Yale School of Medicine, New Haven, Connecticut 06511, USA.,Department of Neuroscience, Yale School of Medicine, New Haven, Connecticut 06510, USA
| | - Eyiyemisi C Damisah
- Department of Neurology, Yale School of Medicine, New Haven, Connecticut 06511, USA.,Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut 06511, USA
| | - Fuyi Chen
- Department of Neurology, Yale School of Medicine, New Haven, Connecticut 06511, USA.,Department of Neuroscience, Yale School of Medicine, New Haven, Connecticut 06510, USA
| | - Alex C Kwan
- Department of Neuroscience, Yale School of Medicine, New Haven, Connecticut 06510, USA.,Department of Psychiatry, Yale School of Medicine, New Haven, Connecticut 06511, USA
| | - Jaime Grutzendler
- Department of Neurology, Yale School of Medicine, New Haven, Connecticut 06511, USA.,Department of Neuroscience, Yale School of Medicine, New Haven, Connecticut 06510, USA
| |
Collapse
|
43
|
Liu W, Klose A, Forman S, Paris ND, Wei-LaPierre L, Cortés-Lopéz M, Tan A, Flaherty M, Miura P, Dirksen RT, Chakkalakal JV. Loss of adult skeletal muscle stem cells drives age-related neuromuscular junction degeneration. eLife 2017; 6. [PMID: 28583253 PMCID: PMC5462534 DOI: 10.7554/elife.26464] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 05/20/2017] [Indexed: 01/04/2023] Open
Abstract
Neuromuscular junction degeneration is a prominent aspect of sarcopenia, the age-associated loss of skeletal muscle integrity. Previously, we showed that muscle stem cells activate and contribute to mouse neuromuscular junction regeneration in response to denervation (Liu et al., 2015). Here, we examined gene expression profiles and neuromuscular junction integrity in aged mouse muscles, and unexpectedly found limited denervation despite a high level of degenerated neuromuscular junctions. Instead, degenerated neuromuscular junctions were associated with reduced contribution from muscle stem cells. Indeed, muscle stem cell depletion was sufficient to induce neuromuscular junction degeneration at a younger age. Conversely, prevention of muscle stem cell and derived myonuclei loss was associated with attenuation of age-related neuromuscular junction degeneration, muscle atrophy, and the promotion of aged muscle force generation. Our observations demonstrate that deficiencies in muscle stem cell fate and post-synaptic myogenesis provide a cellular basis for age-related neuromuscular junction degeneration and associated skeletal muscle decline. DOI:http://dx.doi.org/10.7554/eLife.26464.001
Collapse
Affiliation(s)
- Wenxuan Liu
- Department of Orthopaedics and Rehabilitation, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, United States.,Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, United States
| | - Alanna Klose
- Department of Orthopaedics and Rehabilitation, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, United States
| | - Sophie Forman
- Department of Biology, University of Rochester, Rochester, United States
| | - Nicole D Paris
- Department of Orthopaedics and Rehabilitation, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, United States
| | - Lan Wei-LaPierre
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, United States
| | | | - Aidi Tan
- Bioinformatics Division and Center for Synthetic and Systems Biology, Tsinghua University, Beijing, China.,TNLIST/Department of Automation, Tsinghua University, Beijing, China
| | - Morgan Flaherty
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, United States
| | - Pedro Miura
- Department of Biology, University of Nevada, Reno, United States
| | - Robert T Dirksen
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, United States
| | - Joe V Chakkalakal
- Department of Orthopaedics and Rehabilitation, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, United States.,Stem Cell and Regenerative Medicine Institute, University of Rochester Medical Center, Rochester, United States.,The Rochester Aging Research Center, University of Rochester Medical Center, Rochester, United States
| |
Collapse
|
44
|
Genetic rescue models refute nonautonomous rod cell death in retinitis pigmentosa. Proc Natl Acad Sci U S A 2017; 114:5259-5264. [PMID: 28468800 DOI: 10.1073/pnas.1615394114] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Retinitis pigmentosa (RP) is an inherited neurodegenerative disease, in which the death of mutant rod photoreceptors leads secondarily to the non-cell autonomous death of cone photoreceptors. Gene therapy is a promising treatment strategy. Unfortunately, current methods of gene delivery treat only a fraction of diseased cells, yielding retinas that are a mosaic of treated and untreated rods, as well as cones. In this study, we created two RP mouse models to test whether dying, untreated rods negatively impact treated, rescued rods. In one model, treated and untreated rods were segregated. In the second model, treated and untreated rods were diffusely intermixed, and their ratio was controlled to achieve low-, medium-, or high-efficiency rescue. Analysis of these mosaic retinas demonstrated that rescued rods (and cones) survive, even when they are greatly outnumbered by dying photoreceptors. On the other hand, the rescued photoreceptors did exhibit long-term defects in their outer segments (OSs), which were less severe when more photoreceptors were treated. In summary, our study suggests that even low-efficiency gene therapy may achieve stable survival of rescued photoreceptors in RP patients, albeit with OS dysgenesis.
Collapse
|
45
|
Balasooriya GI, Goschorska M, Piddini E, Rawlins EL. FGFR2 is required for airway basal cell self-renewal and terminal differentiation. Development 2017; 144:1600-1606. [PMID: 28348168 PMCID: PMC5450841 DOI: 10.1242/dev.135681] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Accepted: 03/15/2017] [Indexed: 12/12/2022]
Abstract
Airway stem cells slowly self-renew and produce differentiated progeny to maintain homeostasis throughout the lifespan of an individual. Mutations in the molecular regulators of these processes may drive cancer or degenerative disease, but are also potential therapeutic targets. Conditionally deleting one copy of FGF receptor 2 (FGFR2) in adult mouse airway basal cells results in self-renewal and differentiation phenotypes. We show that FGFR2 signalling correlates with maintenance of expression of a key transcription factor for basal cell self-renewal and differentiation: SOX2. This heterozygous phenotype illustrates that subtle changes in receptor tyrosine kinase signalling can have significant effects, perhaps providing an explanation for the numerous changes seen in cancer. Summary: During adult airway epithelial homeostasis in mice, FGFR2 signalling is required for self-renewing divisions of basal stem cells and to maintain expression of the key transcription factor SOX2.
Collapse
Affiliation(s)
- Gayan I Balasooriya
- Wellcome Trust/CRUK Gurdon Institute, University of Cambridge, Cambridge, CB2 1QN, UK
| | - Maja Goschorska
- Wellcome Trust/CRUK Gurdon Institute, University of Cambridge, Cambridge, CB2 1QN, UK
| | - Eugenia Piddini
- Wellcome Trust/CRUK Gurdon Institute, University of Cambridge, Cambridge, CB2 1QN, UK.,School of Cellular and Molecular Medicine, Faculty of Biomedical Sciences, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, UK
| | - Emma L Rawlins
- Wellcome Trust/CRUK Gurdon Institute, University of Cambridge, Cambridge, CB2 1QN, UK .,Wellcome Trust/MRC Stem Cell Institute and Department of Pathology, University of Cambridge, Cambridge, CB2 1QN, UK
| |
Collapse
|
46
|
Schäfer P, Karl MO. Prospective purification and characterization of Müller glia in the mouse retina regeneration assay. Glia 2017; 65:828-847. [PMID: 28220544 DOI: 10.1002/glia.23130] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 02/01/2017] [Accepted: 02/02/2017] [Indexed: 01/06/2023]
Abstract
Reactive gliosis is an umbrella term for various glia functions in neurodegenerative diseases and upon injury. Specifically, Müller glia (MG) in some species readily regenerate retinal neurons to restore vision loss after insult, whereas mammalian MG respond by reactive gliosis-a heterogeneous response which frequently includes cell hypertrophy and proliferation. Limited regeneration has been stimulated in mammals, with a higher propensity in young MG, and in vitro compared to in vivo, but the underlying processes are unknown. To facilitate studies on the mechanisms regulating and limiting glia functions, we developed a strategy to purify glia and their progeny by fluorescence-activated cell sorting. Dual-transgenic nuclear reporter mice, which label neurons and glia with red and green fluorescent proteins, respectively, have enabled MG enrichment up to 93% purity. We applied this approach to MG in a mouse retina regeneration ex vivo assay. Combined cell size and cell cycle analysis indicates that most MG hypertrophy and a subpopulation proliferates which, over time, become even larger in cell size than the ones that do not proliferate. MG undergo timed differential genomic changes in genes controlling stemness and neurogenic competence; and glial markers are downregulated. Genes that are potentially required for, or associated with, regeneration and reactive gliosis are differentially regulated by retina explant culture time, epidermal growth factor stimulation, and animal age. Thus, MG enrichment facilitates cellular and molecular studies which, in combination with the mouse retina regeneration assay, provide an experimental approach for deciphering mechanisms that possibly regulate reactive gliosis and limit regeneration in mammals.
Collapse
Affiliation(s)
- Patrick Schäfer
- TU Dresden, Center for Regenerative Therapies Dresden (CRTD), Fetscherstr. 107, Dresden, 01307, Germany.,Deutsches Zentrum für Neurodegenerative Erkrankungen e.V. (DZNE), Arnoldstr. 13, Dresden, 01307, Germany
| | - Mike O Karl
- TU Dresden, Center for Regenerative Therapies Dresden (CRTD), Fetscherstr. 107, Dresden, 01307, Germany.,Deutsches Zentrum für Neurodegenerative Erkrankungen e.V. (DZNE), Arnoldstr. 13, Dresden, 01307, Germany
| |
Collapse
|
47
|
Bi P, Yue F, Sato Y, Wirbisky S, Liu W, Shan T, Wen Y, Zhou D, Freeman J, Kuang S. Stage-specific effects of Notch activation during skeletal myogenesis. eLife 2016; 5. [PMID: 27644105 PMCID: PMC5070950 DOI: 10.7554/elife.17355] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 09/17/2016] [Indexed: 12/16/2022] Open
Abstract
Skeletal myogenesis involves sequential activation, proliferation, self-renewal/differentiation and fusion of myogenic stem cells (satellite cells). Notch signaling is known to be essential for the maintenance of satellite cells, but its function in late-stage myogenesis, i.e. post-differentiation myocytes and post-fusion myotubes, is unknown. Using stage-specific Cre alleles, we uncovered distinct roles of Notch1 in mononucleated myocytes and multinucleated myotubes. Specifically, constitutive Notch1 activation dedifferentiates myocytes into Pax7 quiescent satellite cells, leading to severe defects in muscle growth and regeneration, and postnatal lethality. By contrast, myotube-specific Notch1 activation improves the regeneration and exercise performance of aged and dystrophic muscles. Mechanistically, Notch1 activation in myotubes upregulates the expression of Notch ligands, which modulate Notch signaling in the adjacent satellite cells to enhance their regenerative capacity. These results highlight context-dependent effects of Notch activation during myogenesis, and demonstrate that Notch1 activity improves myotube's function as a stem cell niche.
Collapse
Affiliation(s)
- Pengpeng Bi
- Department of Animal Sciences, Purdue University, West Lafayette, United States
| | - Feng Yue
- Department of Animal Sciences, Purdue University, West Lafayette, United States
| | - Yusuke Sato
- Department of Animal Sciences, Purdue University, West Lafayette, United States
| | - Sara Wirbisky
- School of Health Sciences, Purdue University, West Lafayette, United States
| | - Weiyi Liu
- Department of Animal Sciences, Purdue University, West Lafayette, United States
| | - Tizhong Shan
- Department of Animal Sciences, Purdue University, West Lafayette, United States
| | - Yefei Wen
- Department of Animal Sciences, Purdue University, West Lafayette, United States
| | - Daoguo Zhou
- Department of Biological Sciences, Purdue University, West Lafayette, United States
| | - Jennifer Freeman
- School of Health Sciences, Purdue University, West Lafayette, United States.,Center for Cancer Research, Purdue University, West Lafayette, United States
| | - Shihuan Kuang
- Department of Animal Sciences, Purdue University, West Lafayette, United States.,Center for Cancer Research, Purdue University, West Lafayette, United States
| |
Collapse
|
48
|
Richardson GD. Simultaneous Assessment of Cardiomyocyte DNA Synthesis and Ploidy: A Method to Assist Quantification of Cardiomyocyte Regeneration and Turnover. J Vis Exp 2016. [PMID: 27285379 PMCID: PMC4927713 DOI: 10.3791/53979] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Although it is accepted that the heart has a limited potential to regenerate cardiomyocytes following injury and that low levels of cardiomyocyte turnover occur during normal ageing, quantification of these events remains challenging. This is in part due to the rarity of the process and the fact that multiple cellular sources contribute to myocardial maintenance. Furthermore, DNA duplication within cardiomyocytes often leads to a polyploid cardiomyocyte and only rarely leads to new cardiomyocytes by cellular division. In order to accurately quantify cardiomyocyte turnover discrimination between these processes is essential. The protocol described here employs long term nucleoside labeling in order to label all nuclei which have arisen as a result of DNA replication and cardiomyocyte nuclei identified by utilizing nuclei isolation and subsequent PCM1 immunolabeling. Together this allows the accurate and sensitive identification of the nucleoside labeling of the cardiomyocyte nuclei population. Furthermore, 4′,6-diamidino-2-phenylindole labeling and analysis of nuclei ploidy, enables the discrimination of neo-cardiomyocyte nuclei from nuclei which have incorporated nucleoside during polyploidization. Although this method cannot control for cardiomyocyte binucleation, it allows a rapid and robust quantification of neo-cardiomyocyte nuclei while accounting for polyploidization. This method has a number of downstream applications including assessing the potential therapeutics to enhance cardiomyocyte regeneration or investigating the effects of cardiac disease on cardiomyocyte turnover and ploidy. This technique is also compatible with additional downstream immunohistological techniques, allowing quantification of nucleoside incorporation in all cardiac cell types.
Collapse
Affiliation(s)
- Gavin D Richardson
- Institute of Genetic Medicine, International Centre for Life, Newcastle University;
| |
Collapse
|
49
|
Mechanical cell competition kills cells via induction of lethal p53 levels. Nat Commun 2016; 7:11373. [PMID: 27109213 PMCID: PMC4848481 DOI: 10.1038/ncomms11373] [Citation(s) in RCA: 151] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 03/18/2016] [Indexed: 02/07/2023] Open
Abstract
Cell competition is a quality control mechanism that eliminates unfit cells. How cells compete is poorly understood, but it is generally accepted that molecular exchange between cells signals elimination of unfit cells. Here we report an orthogonal mechanism of cell competition, whereby cells compete through mechanical insults. We show that MDCK cells silenced for the polarity gene scribble (scribKD) are hypersensitive to compaction, that interaction with wild-type cells causes their compaction and that crowding is sufficient for scribKD cell elimination. Importantly, we show that elevation of the tumour suppressor p53 is necessary and sufficient for crowding hypersensitivity. Compaction, via activation of Rho-associated kinase (ROCK) and the stress kinase p38, leads to further p53 elevation, causing cell death. Thus, in addition to molecules, cells use mechanical means to compete. Given the involvement of p53, compaction hypersensitivity may be widespread among damaged cells and offers an additional route to eliminate unfit cells. Cell competition is a quality control mechanism to eliminate unfit cells. Here the authors show that physical compaction of less fit cells surrounded by healthy neighbours leads to increased expression of tumour suppressor p53 in the compacted cells, causing cell death.
Collapse
|
50
|
Yang J, Hernandez BJ, Martinez Alanis D, Narvaez del Pilar O, Vila-Ellis L, Akiyama H, Evans SE, Ostrin EJ, Chen J. The development and plasticity of alveolar type 1 cells. Development 2015; 143:54-65. [PMID: 26586225 DOI: 10.1242/dev.130005] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 11/11/2015] [Indexed: 12/16/2022]
Abstract
Alveolar type 1 (AT1) cells cover >95% of the gas exchange surface and are extremely thin to facilitate passive gas diffusion. The development of these highly specialized cells and its coordination with the formation of the honeycomb-like alveolar structure are poorly understood. Using new marker-based stereology and single-cell imaging methods, we show that AT1 cells in the mouse lung form expansive thin cellular extensions via a non-proliferative two-step process while retaining cellular plasticity. In the flattening step, AT1 cells undergo molecular specification and remodel cell junctions while remaining connected to their epithelial neighbors. In the folding step, AT1 cells increase in size by more than 10-fold and undergo cellular morphogenesis that matches capillary and secondary septa formation, resulting in a single AT1 cell spanning multiple alveoli. Furthermore, AT1 cells are an unexpected source of VEGFA and their normal development is required for alveolar angiogenesis. Notably, a majority of AT1 cells proliferate upon ectopic SOX2 expression and undergo stage-dependent cell fate reprogramming. These results provide evidence that AT1 cells have both structural and signaling roles in alveolar maturation and can exit their terminally differentiated non-proliferative state. Our findings suggest that AT1 cells might be a new target in the pathogenesis and treatment of lung diseases associated with premature birth.
Collapse
Affiliation(s)
- Jun Yang
- Department of Pulmonary Medicine, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Belinda J Hernandez
- Department of Pulmonary Medicine, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Denise Martinez Alanis
- Department of Pulmonary Medicine, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | | | - Lisandra Vila-Ellis
- Department of Pulmonary Medicine, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA Tecnologico de Monterrey - Escuela de Medicina, Monterrey 64710, Mexico
| | - Haruhiko Akiyama
- Department of Orthopedics, Kyoto University, Sakyo, Kyoto 606-8507, Japan
| | - Scott E Evans
- Department of Pulmonary Medicine, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Edwin J Ostrin
- Department of Pulmonary Medicine, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Jichao Chen
- Department of Pulmonary Medicine, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA Center for Stem Cells and Developmental Biology, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|