1
|
Wang J, Dong X, Lei J, Zhang Y, Chen S, He Y. β-catenin Orchestrates Gli1+ Cell Fate in Condylar Development and TMJOA. J Dent Res 2024; 103:1291-1301. [PMID: 39400124 DOI: 10.1177/00220345241274354] [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] [Indexed: 10/15/2024] Open
Abstract
The fibrocartilage stem cells (FCSCs) on the surface of the condyle play an essential role in cartilage homeostasis and regeneration. However, few well-defined stem cell markers have been identified for the analysis of FCSCs' cell fate and regulation mechanism. In this study, we first mapped the transcriptional landscape of the condylar cartilage and identified a Gli1+ subset. Label-retaining cells and our lineage-tracing study showed that Gli1 labeled a group of FCSCs. Conditional knockout β-catenin inhibited Gli1+ cells differentiating into hypertrophic chondrocytes. In discectomy-induced temporomandibular joint osteoarthritis (TMJOA), Gli1+ cells were further activated, and their differentiation into hypertrophic chondrocytes was accelerated, which induced stem cell pool depletion. The deletion of β-catenin in Gli1+ cells preserved the FCSC pool and alleviated TMJOA cartilage degeneration. Collectively, we uncovered that a Gli1+ FCSC subpopulation and Wnt/β-catenin signaling orchestrate the Gli1+ cell fate in condyle postnatal development and TMJOA.
Collapse
Affiliation(s)
- J Wang
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - X Dong
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - J Lei
- Center for TMD and Orofacial Pain, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Y Zhang
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - S Chen
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Y He
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| |
Collapse
|
2
|
Deng Y, He Y, Xu J, He H, Zhang M, Li G. Cardiac Fibroblasts regulate myocardium and coronary vasculature development via the collagen signaling pathway. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.11.612512. [PMID: 39314489 PMCID: PMC11418987 DOI: 10.1101/2024.09.11.612512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
The fibroblast (FB), cardiomyocyte (CM), and vascular endothelial cell (Vas_EC) are the three major cell types in the heart, yet their relationships during development are largely unexplored. To address this gap, we employed RNA staining of the FB marker gene Col1a1 together with the CM marker gene Actn2 and the Vas_EC marker gene Cdh5 at different stages. This approach enabled us to discern the anatomical pattern of cardiac FBs and identify approximately one EC and four CMs directly interacting with each FB. Molecularly, through the analysis of single-cell mRNA sequencing (scRNA-seq) data, we unveiled collagen as the top signaling molecule derived from FBs influencing CM and Vas_EC development. Subsequently, we used a Pdgfra-CreER controlled diphtheria toxin A (DTA) system to ablate the FBs at different stages. We found that the ablation of FBs disrupted myocardium and vasculature development and led to embryonic heart defects. Using scRNA-seq, we further profiled the ablated hearts and identified molecular defects in their ventricular CMs and Vas_ECs compared to control hearts. Moreover, we identified a reduction of collagen in the ablated hearts and predicted collagen as the major signaling pathway regulating the differentially expressed genes in the ablated ventricular CMs. Finally, we performed both short-term and long-term fibroblast ablation at the neonatal stage. We found that short-term ablation caused a reduction in collagen and Vas_EC density, while long-term ablation may induce compensatory collagen expression without causing heart function reduction. In summary, our study has identified the function of fibroblasts in regulating myocardium and vasculature development and implicated an important role for the collagen pathway in this process.
Collapse
Affiliation(s)
- Yiting Deng
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201, USA
| | - Yuanhang He
- Tsinghua University, Tsinghua medicine, School of Medicine, Beijing, China
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201, USA
| | - Juan Xu
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201, USA
| | - Haoting He
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201, USA
| | - Manling Zhang
- Vascular Medicine Institute, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Guang Li
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201, USA
| |
Collapse
|
3
|
Gordon RA, Cosgrove HA, Marinov A, Gingras S, Tilstra JS, Campbell AM, Bastacky SI, Kashgarian M, Perl A, Nickerson KM, Shlomchik MJ. NADPH oxidase in B cells and macrophages protects against murine lupus by regulation of TLR7. JCI Insight 2024; 9:e178563. [PMID: 39042716 PMCID: PMC11343599 DOI: 10.1172/jci.insight.178563] [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: 12/15/2023] [Accepted: 07/12/2024] [Indexed: 07/25/2024] Open
Abstract
Loss of NADPH oxidase (NOX2) exacerbates systemic lupus erythematosus (SLE) in mice and humans, but the mechanisms underlying this effect remain unclear. To identify the cell lineages in which NOX2 deficiency drives SLE, we employed conditional KO and chimeric approaches to delete Cybb in several hematopoietic cell lineages of MRL.Faslpr SLE-prone mice. Deletion of Cybb in macrophages/monocytes exacerbated SLE nephritis, though not to the degree observed in the Cybb global KOs. Unexpectedly, the absence of Cybb in B cells resulted in profound glomerulonephritis and interstitial nephritis, rivaling that seen with global deletion. Furthermore, we identified that NOX2 is a key regulator of TLR7, a driver of SLE pathology, both globally and specifically in B cells. This is mediated in part through suppression of TLR7-mediated NF-κB signaling in B cells. Thus, NOX2's immunomodulatory effect in SLE is orchestrated not only by its function in the myeloid compartment, but through a pivotal role in B cells by selectively inhibiting TLR7 signaling.
Collapse
Affiliation(s)
- Rachael A. Gordon
- Department of Immunology and
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Haylee A. Cosgrove
- Department of Immunology and
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | | | | | - Jeremy S. Tilstra
- Department of Immunology and
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Allison M. Campbell
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Sheldon I. Bastacky
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Michael Kashgarian
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Andras Perl
- Departments of Medicine, Microbiology and Immunology, Biochemistry and Molecular Biology, State University of New York, Upstate Medical University, College of Medicine, Syracuse, New York, USA
| | | | | |
Collapse
|
4
|
Higham JP, Bhebhe CN, Gupta RA, Tranter MM, Barakat FM, Dogra H, Bab N, Wozniak E, Barker KH, Wilson CH, Mein CA, Raine T, Cox JJ, Wood JN, Croft NM, Wright PD, Bulmer DC. Transcriptomic profiling reveals a pronociceptive role for angiotensin II in inflammatory bowel disease. Pain 2024; 165:1592-1604. [PMID: 38293826 PMCID: PMC11190897 DOI: 10.1097/j.pain.0000000000003159] [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: 07/26/2023] [Revised: 10/20/2023] [Accepted: 10/29/2023] [Indexed: 02/01/2024]
Abstract
ABSTRACT Visceral pain is a leading cause of morbidity in inflammatory bowel disease (IBD), contributing significantly to reduced quality of life. Currently available analgesics often lack efficacy or have intolerable side effects, driving the need for a more complete understanding of the mechanisms causing pain. Whole transcriptome gene expression analysis was performed by bulk RNA sequencing of colonic biopsies from patients with ulcerative colitis (UC) and Crohn's disease (CD) reporting abdominal pain and compared with noninflamed control biopsies. Potential pronociceptive mediators were identified based on gene upregulation in IBD biopsy tissue and cognate receptor expression in murine colonic sensory neurons. Pronociceptive activity of identified mediators was assessed in assays of sensory neuron and colonic afferent activity. RNA sequencing analysis highlighted a 7.6-fold increase in the expression of angiotensinogen transcripts, Agt , which encode the precursor to angiotensin II (Ang II), in samples from UC patients ( P = 3.2 × 10 -8 ). Consistent with the marked expression of the angiotensin AT 1 receptor in colonic sensory neurons, Ang II elicited an increase in intracellular Ca 2+ in capsaicin-sensitive, voltage-gated sodium channel subtype Na V 1.8-positive sensory neurons. Ang II also evoked action potential discharge in high-threshold colonic nociceptors. These effects were inhibited by the AT 1 receptor antagonist valsartan. Findings from our study identify AT 1 receptor-mediated colonic nociceptor activation as a novel pathway of visceral nociception in patients with UC. This work highlights the potential utility of angiotensin receptor blockers, such as valsartan, as treatments for pain in IBD.
Collapse
Affiliation(s)
- James P. Higham
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| | - Charity N. Bhebhe
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| | - Rohit A. Gupta
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| | - Michael M. Tranter
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Farah M. Barakat
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Harween Dogra
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Natalie Bab
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Eva Wozniak
- Genome Centre, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Katie H. Barker
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| | - Catherine H. Wilson
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| | - Charles A. Mein
- Genome Centre, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Tim Raine
- Department of Gastroenterology, Addenbrookes Hospital, Cambridge University Teaching Hospitals, Cambridge, United Kingdom
| | - James J. Cox
- Wolfson Institute for Biomedical Research, University College London, London, United Kingdom
| | - John N. Wood
- Wolfson Institute for Biomedical Research, University College London, London, United Kingdom
| | - Nicholas M. Croft
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Paul D. Wright
- LifeArc, SBC Open Innovation Campus, Stevenage, United Kingdom
| | - David C. Bulmer
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
5
|
Yang N, Sun Y, Han B, Deng N, Li G, Han Q, Wang Y, Cai H, Liu F, Cao B, Deng W, Bao H, Kong S, Lu J, Wang H. Trophoblastic signals facilitate endometrial interferon response and lipid metabolism, ensuring normal decidualization. Cell Rep 2024; 43:114246. [PMID: 38762885 DOI: 10.1016/j.celrep.2024.114246] [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: 11/03/2023] [Revised: 04/04/2024] [Accepted: 05/02/2024] [Indexed: 05/21/2024] Open
Abstract
The decidua plays a crucial role in providing structural and trophic support to the developing conceptus before placentation. Following embryo attachment, embryonic components intimately interact with the decidual tissue. While evidence indicates the participation of embryo-derived factors in crosstalk with the uterus, the extent of their impact on post-implantation decidual development requires further investigation. Here, we utilize transgenic mouse models to selectively eliminate primary trophoblast giant cells (pTGCs), the embryonic cells that interface with maternal tissue at the forefront. pTGC ablation impairs decidualization and compromises decidual interferon response and lipid metabolism. Mechanistically, pTGCs release factors such as interferon kappa (IFNK) to strengthen the decidual interferon response and lipoprotein lipase (LPL) to enhance lipid accumulation within the decidua, thereby promoting decidualization. This study presents genetic and metabolomic evidence reinforcing the proactive role of pTGC-derived factors in mobilizing maternal resources to strengthen decidualization, facilitating the normal progression of early pregnancy.
Collapse
Affiliation(s)
- Ningjie Yang
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Yang Sun
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Bing Han
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Na Deng
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Gaizhen Li
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Qian Han
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Yinan Wang
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Han Cai
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Fan Liu
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China; State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Bin Cao
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Wenbo Deng
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Haili Bao
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China.
| | - Shuangbo Kong
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China.
| | - Jinhua Lu
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China.
| | - Haibin Wang
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China; State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China.
| |
Collapse
|
6
|
Maraslioglu-Sperber A, Blanc F, Heller S. Murine cochlear damage models in the context of hair cell regeneration research. Hear Res 2024; 447:109021. [PMID: 38703432 DOI: 10.1016/j.heares.2024.109021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 04/16/2024] [Accepted: 04/26/2024] [Indexed: 05/06/2024]
Abstract
Understanding the complex pathologies associated with hearing loss is a significant motivation for conducting inner ear research. Lifelong exposure to loud noise, ototoxic drugs, genetic diversity, sex, and aging collectively contribute to human hearing loss. Replicating this pathology in research animals is challenging because hearing impairment has varied causes and different manifestations. A central aspect, however, is the loss of sensory hair cells and the inability of the mammalian cochlea to replace them. Researching therapeutic strategies to rekindle regenerative cochlear capacity, therefore, requires the generation of animal models in which cochlear hair cells are eliminated. This review discusses different approaches to ablate cochlear hair cells in adult mice. We inventoried the cochlear cyto- and histo-pathology caused by acoustic overstimulation, systemic and locally applied drugs, and various genetic tools. The focus is not to prescribe a perfect damage model but to highlight the limitations and advantages of existing approaches and identify areas for further refinement of damage models for use in regenerative studies.
Collapse
Affiliation(s)
- Ayse Maraslioglu-Sperber
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Fabian Blanc
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Otolaryngology - Head & Neck Surgery, University Hospital Gui de Chauliac, University of Montpellier, Montpellier, France
| | - Stefan Heller
- Department of Otolaryngology - Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
7
|
Kuriki M, Korb A, Comai G, Tajbakhsh S. Interplay between Pitx2 and Pax7 temporally governs specification of extraocular muscle stem cells. PLoS Genet 2024; 20:e1010935. [PMID: 38875306 PMCID: PMC11178213 DOI: 10.1371/journal.pgen.1010935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 03/05/2024] [Indexed: 06/16/2024] Open
Abstract
Gene regulatory networks that act upstream of skeletal muscle fate determinants are distinct in different anatomical locations. Despite recent efforts, a clear understanding of the cascade of events underlying the emergence and maintenance of the stem cell pool in specific muscle groups remains unresolved and debated. Here, we invalidated Pitx2 with multiple Cre-driver mice prenatally, postnatally, and during lineage progression. We showed that this gene becomes progressively dispensable for specification and maintenance of the muscle stem (MuSC) cell pool in extraocular muscles (EOMs) despite being, together with Myf5, a major upstream regulator during early development. Moreover, constitutive inactivation of Pax7 postnatally led to a greater loss of MuSCs in the EOMs compared to the limb. Thus, we propose a relay between Pitx2, Myf5 and Pax7 for EOM stem cell maintenance. We demonstrate also that MuSCs in the EOMs adopt a quiescent state earlier that those in limb muscles and do not spontaneously proliferate in the adult, yet EOMs have a significantly higher content of Pax7+ MuSCs per area pre- and post-natally. Finally, while limb MuSCs proliferate in the mdx mouse model for Duchenne muscular dystrophy, significantly less MuSCs were present in the EOMs of the mdx mouse model compared to controls, and they were not proliferative. Overall, our study provides a comprehensive in vivo characterisation of MuSC heterogeneity along the body axis and brings further insights into the unusual sparing of EOMs during muscular dystrophy.
Collapse
Affiliation(s)
- Mao Kuriki
- Institut Pasteur, Université Paris Cité, CNRS UMR 3738, Stem Cells & Development Unit, Institut Pasteur, Paris, France
| | - Amaury Korb
- Institut Pasteur, Université Paris Cité, CNRS UMR 3738, Stem Cells & Development Unit, Institut Pasteur, Paris, France
| | - Glenda Comai
- Institut Pasteur, Université Paris Cité, CNRS UMR 3738, Stem Cells & Development Unit, Institut Pasteur, Paris, France
| | - Shahragim Tajbakhsh
- Institut Pasteur, Université Paris Cité, CNRS UMR 3738, Stem Cells & Development Unit, Institut Pasteur, Paris, France
| |
Collapse
|
8
|
Lee D, Benvie AM, Steiner BM, Kolba NJ, Ford JG, McCabe SM, Jiang Y, Berry DC. Smooth muscle cell-derived Cxcl12 directs macrophage accrual and sympathetic innervation to control thermogenic adipose tissue. Cell Rep 2024; 43:114169. [PMID: 38678562 PMCID: PMC11413973 DOI: 10.1016/j.celrep.2024.114169] [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: 03/22/2024] [Revised: 04/11/2024] [Accepted: 04/15/2024] [Indexed: 05/01/2024] Open
Abstract
Sympathetic innervation of brown adipose tissue (BAT) controls mammalian adaptative thermogenesis. However, the cellular and molecular underpinnings contributing to BAT innervation remain poorly defined. Here, we show that smooth muscle cells (SMCs) support BAT growth, lipid utilization, and thermogenic plasticity. Moreover, we find that BAT SMCs express and control the bioavailability of Cxcl12. SMC deletion of Cxcl12 fosters brown adipocyte lipid accumulation, reduces energy expenditure, and increases susceptibility to diet-induced metabolic dysfunction. Mechanistically, we find that Cxcl12 stimulates CD301+ macrophage recruitment and supports sympathetic neuronal maintenance. Administering recombinant Cxcl12 to obese mice or leptin-deficient (Ob/Ob) mice is sufficient to boost macrophage presence and drive sympathetic innervation to restore BAT morphology and thermogenic responses. Altogether, our data reveal an SMC chemokine-dependent pathway linking immunological infiltration and sympathetic innervation as a rheostat for BAT maintenance and thermogenesis.
Collapse
Affiliation(s)
- Derek Lee
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Abigail M Benvie
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Benjamin M Steiner
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Nikolai J Kolba
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Josie G Ford
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Sean M McCabe
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Yuwei Jiang
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Daniel C Berry
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
9
|
Xu J, Deng Y, Li G. Keratin 19 (Krt19) is a novel marker gene for epicardial cells. Front Genet 2024; 15:1385867. [PMID: 38831775 PMCID: PMC11145414 DOI: 10.3389/fgene.2024.1385867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 04/26/2024] [Indexed: 06/05/2024] Open
Abstract
Epicardial cells regulate heart growth by secreting numerous growth factors and undergoing lineage specification into other cardiac lineages. However, the lack of specific marker genes for epicardial cells has hindered the understanding of this cell type in heart development. Through the analysis of a cardiac single cell mRNA sequencing dataset, we identified a novel epicardial gene named Keratin 19 (Krt19). Further analysis of the expression patterns of Krt19 and Wt1, a well-known epicardial gene, revealed their preferences in major cardiac cell types. Using lineage-tracing analysis, we analyzed Krt19-CreER labeled cells at multiple time windows and found that it labels epicardial cells at both embryonic and neonatal stages. Furthermore, we studied the function of epicardial cells using a diphtheria toxin A chain (DTA)-based cell ablation system. We discovered that Krt19-CreER labeled cells are essential for fetal heart development. Finally, we investigated the function of Krt19-CreER and Wt1-CreER labeled cells in neonatal mouse development. We observed that the Krt19-CreER; Rosa-DTA mice displayed a smaller size after tamoxifen treatment, suggesting the potential importance of Krt19-CreER labeled cells in neonatal mouse development. Additionally, we found that Wt1-CreER; Rosa-DTA mice died at early stages, likely due to defects in the kidney and spleen. In summary, we have identified Krt19 as a new epicardial cell marker gene and further explored the function of epicardial cells using the Krt19-CreER and Wt1-CreER-mediated DTA ablation system.
Collapse
Affiliation(s)
| | | | - Guang Li
- Department of Cell Biology, Center for Integrative Organ Systems, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
10
|
Lim JJY, Murata Y, Yuri S, Kitamuro K, Kawai T, Isotani A. Generating an organ-deficient animal model using a multi-targeted CRISPR-Cas9 system. Sci Rep 2024; 14:10636. [PMID: 38724644 PMCID: PMC11082136 DOI: 10.1038/s41598-024-61167-3] [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: 09/12/2023] [Accepted: 05/02/2024] [Indexed: 05/12/2024] Open
Abstract
Gene-knockout animal models with organ-deficient phenotypes used for blastocyst complementation are generally not viable. Animals need to be maintained as heterozygous mutants, and homozygous mutant embryos yield only one-fourth of all embryos. In this study, we generated organ-deficient embryos using the CRISPR-Cas9-sgRNAms system that induces cell death with a single-guide RNA (sgRNAms) targeting multiple sites in the genome. The Cas9-sgRNAms system interrupted cell proliferation and induced cell ablation in vitro. The mouse model had Cas9 driven by the Foxn1 promoter with a ubiquitous expression cassette of sgRNAms at the Rosa26 locus (Foxn1Cas9; Rosa26_ms). It showed an athymic phenotype similar to that of nude mice but was not hairless. Eventually, a rat cell-derived thymus in an interspecies chimera was generated by blastocyst complementation of Foxn1Cas9; Rosa26_ms mouse embryos with rat embryonic stem cells. Theoretically, a half of the total embryos has the Cas9-sgRNAms system because Rosa26_ms could be maintained as homozygous.
Collapse
Affiliation(s)
- Jonathan Jun-Yong Lim
- Laboratory of Organ Developmental Engineering, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5 Takayama-cho, Ikoma, Nara, 630-0912, Japan
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore City, Singapore
| | - Yamato Murata
- Laboratory of Organ Developmental Engineering, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5 Takayama-cho, Ikoma, Nara, 630-0912, Japan
| | - Shunsuke Yuri
- Laboratory of Organ Developmental Engineering, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5 Takayama-cho, Ikoma, Nara, 630-0912, Japan
| | - Kohei Kitamuro
- Laboratory of Organ Developmental Engineering, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5 Takayama-cho, Ikoma, Nara, 630-0912, Japan
| | - Taro Kawai
- Laboratory of Molecular Immunobiology, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5 Takayama-cho, Ikoma, Nara, 630-0912, Japan
- Life Science Collaboration Center (LiSCo), Nara Institute of Science and Technology, 8916-5 Takayama-cho, Ikoma, Nara, 630-0192, Japan
| | - Ayako Isotani
- Laboratory of Organ Developmental Engineering, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5 Takayama-cho, Ikoma, Nara, 630-0912, Japan.
- Life Science Collaboration Center (LiSCo), Nara Institute of Science and Technology, 8916-5 Takayama-cho, Ikoma, Nara, 630-0192, Japan.
| |
Collapse
|
11
|
Su J, Byer L, Liang Y, Fox MA. Distribution, development, and identity of retinal ganglion cells labeled in the Sert-Cre reporter mouse. J Comp Neurol 2024; 532:e25606. [PMID: 38544361 DOI: 10.1002/cne.25606] [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: 05/25/2023] [Revised: 02/19/2024] [Accepted: 03/07/2024] [Indexed: 05/01/2024]
Abstract
The mouse retina contains over 40 types of retinal ganglion cells (RGCs) that differ in morphology, function, or gene expression. RGCs also differ by whether their axons target the brain.s ipsilateral or contralateral hemisphere. Contralaterally projecting RGCs (contraRGCs) are widespread in mouse retina, whereas ipsilateral projecting RGCs (ipsiRGCs) are confined to the ventro-temporal (VT) crescent of retina. In this study, we employed the Sert-Cre transgenic line, which had been reported to selectively label ipsiRGCs, to study ipsiRGCs during development. Although the number of Cre-expressing ipsiRGCs did not significantly increase with postnatal age, the region of retina that they occupied did, and by adulthood represented ~30% of the retinal surface. Unexpectedly, genetic ablation of Sert-Cre cells failed to fully disrupt ipsilateral projecting retinal axons, suggesting that not all ipsiRGCs generated Cre in Sert-Cre mice. To test this hypothesis, we retrogradely labeled ipsiRGCs in Sert-Cre mice which revealed that not all ipsiRGCs are labeled in Sert-Cre mice and a small population of contraRGCs flanking the VT crescent generates Cre in this line. These results do not negate the usefulness of the Sert-Cre mouse but do raise important caveats to the interpretation of such studies.
Collapse
Affiliation(s)
- Jianmin Su
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, USA
- School of Neuroscience, College of Science, Virginia Tech, Blacksburg, Virginia, USA
| | - Lillian Byer
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, USA
- Department of Neuroscience, Davidson College, Davidson, North Carolina, USA
- NeuroSURF Program, Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, USA
| | - Yanping Liang
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, USA
| | - Michael A Fox
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, USA
- School of Neuroscience, College of Science, Virginia Tech, Blacksburg, Virginia, USA
- Department of Biological Sciences, College of Science, Virginia Tech, Blacksburg, Virginia, USA
- Department of Biology, College of Natural Sciences, University of Massachusetts Amherst, Amherst, Massachusetts, USA
| |
Collapse
|
12
|
Huang JL, Pourhosseinzadeh MS, Lee S, Krämer N, Guillen JV, Cinque NH, Aniceto P, Momen AT, Koike S, Huising MO. Paracrine signalling by pancreatic δ cells determines the glycaemic set point in mice. Nat Metab 2024; 6:61-77. [PMID: 38195859 PMCID: PMC10919447 DOI: 10.1038/s42255-023-00944-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 11/09/2023] [Indexed: 01/11/2024]
Abstract
While pancreatic β and α cells are considered the main drivers of blood glucose homeostasis through insulin and glucagon secretion, the contribution of δ cells and somatostatin (SST) secretion to glucose homeostasis remains unresolved. Here we provide a quantitative assessment of the physiological contribution of δ cells to the glycaemic set point in mice. Employing three orthogonal mouse models to remove SST signalling within the pancreas or transplanted islets, we demonstrate that ablating δ cells or SST leads to a sustained decrease in the glycaemic set point. This reduction coincides with a decreased glucose threshold for insulin response from β cells, leading to increased insulin secretion to the same glucose challenge. Our data demonstrate that β cells are sufficient to maintain stable glycaemia and reveal that the physiological role of δ cells is to provide tonic feedback inhibition that reduces the β cell glucose threshold and consequently lowers the glycaemic set point in vivo.
Collapse
Affiliation(s)
- Jessica L Huang
- Department of Neurobiology, Physiology and Behavior, College of Biological Sciences, University of California, Davis, CA, USA
| | - Mohammad S Pourhosseinzadeh
- Department of Neurobiology, Physiology and Behavior, College of Biological Sciences, University of California, Davis, CA, USA
| | - Sharon Lee
- Department of Neurobiology, Physiology and Behavior, College of Biological Sciences, University of California, Davis, CA, USA
| | - Niels Krämer
- Department of Neurobiology, Physiology and Behavior, College of Biological Sciences, University of California, Davis, CA, USA
- Department of Animal Ecology and Physiology, Radboud Institute for Biological and Environmental Sciences, Radboud University, Nijmegen, The Netherlands
| | - Jaresley V Guillen
- Department of Neurobiology, Physiology and Behavior, College of Biological Sciences, University of California, Davis, CA, USA
| | - Naomi H Cinque
- Department of Neurobiology, Physiology and Behavior, College of Biological Sciences, University of California, Davis, CA, USA
| | - Paola Aniceto
- Department of Neurobiology, Physiology and Behavior, College of Biological Sciences, University of California, Davis, CA, USA
| | - Ariana T Momen
- Department of Neurobiology, Physiology and Behavior, College of Biological Sciences, University of California, Davis, CA, USA
| | - Shinichiro Koike
- Department of Nutrition, University of California, Davis, CA, USA
| | - Mark O Huising
- Department of Neurobiology, Physiology and Behavior, College of Biological Sciences, University of California, Davis, CA, USA.
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, CA, USA.
| |
Collapse
|
13
|
Teske NC, Dyckhoff-Shen S, Beckenbauer P, Bewersdorf JP, Engelen-Lee JY, Hammerschmidt S, Kälin RE, Pfister HW, Brouwer MC, Klein M, Glass R, van de Beek D, Koedel U. Pericytes are protective in experimental pneumococcal meningitis through regulating leukocyte infiltration and blood-brain barrier function. J Neuroinflammation 2023; 20:267. [PMID: 37978545 PMCID: PMC10655320 DOI: 10.1186/s12974-023-02938-z] [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: 07/16/2023] [Accepted: 10/27/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND Brain pericytes participate in the regulation of cerebral blood flow and the maintenance of blood-brain barrier integrity. Because of their perivascular localization, their receptor repertoire, and their potential ability to respond to inflammatory and infectious stimuli by producing various cytokines and chemokines, these cells are also thought to play an active role in the immune response to brain infections. This assumption is mainly supported by in vitro studies, investigations in in vivo disease models are largely missing. Here, we analysed the role of brain pericytes in pneumococcal meningitis, in vitro and in vivo in two animal models of pneumococcal meningitis. METHODS Primary murine and human pericytes were stimulated with increasing concentrations of different serotypes of Streptococcus pneumoniae in the presence or absence of Toll-like receptor inhibitors and their cell viability and cytokine production were monitored. To gain insight into the role of pericytes in brain infection in vivo, we performed studies in a zebrafish embryo model of pneumococcal meningitis in which pericytes were pharmacologically depleted. Furthermore, we analyzed the impact of genetically induced pericyte ablation on disease progression, intracranial complications, and brain inflammation in an adult mouse model of this disease. RESULTS Both murine and human pericytes reacted to pneumococcal exposure with the release of selected cytokines. This cytokine release is pneumolysin-dependent, TLR-dependent in murine (but not human) pericytes and can be significantly increased by macrophage-derived IL-1b. Pharmacological depletion of pericytes in zebrafish embryos resulted in increased cerebral edema and mortality due to pneumococcal meningitis. Correspondingly, in an adult mouse meningitis model, a more pronounced blood-brain barrier disruption and leukocyte infiltration, resulting in an unfavorable disease course, was observed following genetic pericyte ablation. The degree of leukocyte infiltration positively correlated with an upregulation of chemokine expression in the brains of pericyte-depleted mice. CONCLUSIONS Our findings show that pericytes play a protective role in pneumococcal meningitis by impeding leukocyte migration and preventing blood-brain barrier breaching. Thus, preserving the integrity of the pericyte population has the potential as a new therapeutic strategy in pneumococcal meningitis.
Collapse
Affiliation(s)
- Nina C Teske
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany.
- ESCMID Study Group for Infections of the Brain, Basel, Switzerland.
- Department of Neurology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands.
- Department of Neurosurgery, LMU University Hospital, LMU Munich, Munich, Germany.
| | | | - Paul Beckenbauer
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| | | | - Joo-Yeon Engelen-Lee
- ESCMID Study Group for Infections of the Brain, Basel, Switzerland
- Department of Neurology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Sven Hammerschmidt
- Department Genetics of Microorganisms, Interfaculty Institute of Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Roland E Kälin
- Neurosurgical Research, Department of Neurosurgery, LMU University Hospital, LMU Munich, Munich, Germany
- Walter Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Hans-Walter Pfister
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
- ESCMID Study Group for Infections of the Brain, Basel, Switzerland
| | - Matthijs C Brouwer
- ESCMID Study Group for Infections of the Brain, Basel, Switzerland
- Department of Neurology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Matthias Klein
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
- ESCMID Study Group for Infections of the Brain, Basel, Switzerland
| | - Rainer Glass
- Neurosurgical Research, Department of Neurosurgery, LMU University Hospital, LMU Munich, Munich, Germany
| | - Diederik van de Beek
- ESCMID Study Group for Infections of the Brain, Basel, Switzerland
- Department of Neurology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Uwe Koedel
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
- ESCMID Study Group for Infections of the Brain, Basel, Switzerland
| |
Collapse
|
14
|
Konkimalla A, Konishi S, Macadlo L, Kobayashi Y, Farino ZJ, Miyashita N, El Haddad L, Morowitz J, Barkauskas CE, Agarwal P, Souma T, ElMallah MK, Tata A, Tata PR. Transitional cell states sculpt tissue topology during lung regeneration. Cell Stem Cell 2023; 30:1486-1502.e9. [PMID: 37922879 PMCID: PMC10762634 DOI: 10.1016/j.stem.2023.10.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 06/22/2023] [Accepted: 10/03/2023] [Indexed: 11/07/2023]
Abstract
Organ regeneration requires dynamic cell interactions to reestablish cell numbers and tissue architecture. While we know the identity of progenitor cells that replace lost tissue, the transient states they give rise to and their role in repair remain elusive. Here, using multiple injury models, we find that alveolar fibroblasts acquire distinct states marked by Sfrp1 and Runx1 that influence tissue remodeling and reorganization. Unexpectedly, ablation of alveolar epithelial type-1 (AT1) cells alone is sufficient to induce tissue remodeling and transitional states. Integrated scRNA-seq followed by genetic interrogation reveals RUNX1 is a key driver of fibroblast states. Importantly, the ectopic induction or accumulation of epithelial transitional states induce rapid formation of transient alveolar fibroblasts, leading to organ-wide fibrosis. Conversely, the elimination of epithelial or fibroblast transitional states or RUNX1 loss, leads to tissue simplification resembling emphysema. This work uncovered a key role for transitional states in orchestrating tissue topologies during regeneration.
Collapse
Affiliation(s)
- Arvind Konkimalla
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA; Medical Scientist Training Program, Duke University School of Medicine, Durham, NC 27710, USA
| | - Satoshi Konishi
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Lauren Macadlo
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Yoshihiko Kobayashi
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Zachary J Farino
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Naoya Miyashita
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Léa El Haddad
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, School of Medicine, Duke University, Durham, NC, USA
| | - Jeremy Morowitz
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Christina E Barkauskas
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Pankaj Agarwal
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Tomokazu Souma
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA; Duke Regeneration Center, Duke University, Durham, NC 27710, USA
| | - Mai K ElMallah
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, School of Medicine, Duke University, Durham, NC, USA
| | - Aleksandra Tata
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA.
| | - Purushothama Rao Tata
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA; Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA; Duke Regeneration Center, Duke University, Durham, NC 27710, USA; Center for Advanced Genomic Technologies, Duke University, Durham, NC 27710, USA; Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710, USA.
| |
Collapse
|
15
|
Fuller AM, Luiz A, Tian N, Arcangeletti M, Iseppon F, Sexton JE, Millet Q, Caxaria S, Ketabi N, Celik P, Wood JN, Sikandar S. Gate control of sensory neurotransmission in peripheral ganglia by proprioceptive sensory neurons. Brain 2023; 146:4033-4039. [PMID: 37249190 PMCID: PMC10549771 DOI: 10.1093/brain/awad182] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 04/12/2023] [Accepted: 05/18/2023] [Indexed: 05/31/2023] Open
Abstract
Melzak and Wall's gate control theory proposed that innocuous input into the dorsal horn of the spinal cord represses pain-inducing nociceptive input. Here we show that input from proprioceptive parvalbumin-expressing sensory neurons tonically represses nociceptor activation within dorsal root ganglia. Deletion of parvalbumin-positive sensory neurons leads to enhanced nociceptor activity measured with GCaMP3, increased input into wide dynamic range neurons of the spinal cord and increased acute and spontaneous pain behaviour, as well as potentiated innocuous sensation. Parvalbumin-positive sensory neurons express the enzymes and transporters necessary to produce vesicular GABA that is known to be released from depolarized somata. These observations support the view that gate control mechanisms occur peripherally within dorsal root ganglia.
Collapse
Affiliation(s)
- Alice M Fuller
- William Harvey Research Institute, Queen Mary University of London, London EC1M 6BQ, UK
- Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
| | - Ana Luiz
- Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
| | - Naxi Tian
- Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
| | - Manuel Arcangeletti
- Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
| | - Federico Iseppon
- Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
| | - Jane E Sexton
- Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
| | - Queensta Millet
- Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
| | - Sara Caxaria
- William Harvey Research Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Niloofar Ketabi
- William Harvey Research Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - Petek Celik
- William Harvey Research Institute, Queen Mary University of London, London EC1M 6BQ, UK
| | - John N Wood
- Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK
| | - Shafaq Sikandar
- William Harvey Research Institute, Queen Mary University of London, London EC1M 6BQ, UK
| |
Collapse
|
16
|
Robinson KS, Toh GA, Firdaus MJ, Tham KC, Rozario P, Lim CK, Toh YX, Lau ZH, Binder SC, Mayer J, Bonnard C, Schmidt FI, Common JE, Zhong FL. Diphtheria toxin activates ribotoxic stress and NLRP1 inflammasome-driven pyroptosis. J Exp Med 2023; 220:e20230105. [PMID: 37642997 PMCID: PMC10465786 DOI: 10.1084/jem.20230105] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 06/01/2023] [Accepted: 07/14/2023] [Indexed: 08/31/2023] Open
Abstract
The ZAKα-driven ribotoxic stress response (RSR) is activated by ribosome stalling and/or collisions. Recent work demonstrates that RSR also plays a role in innate immunity by activating the human NLRP1 inflammasome. Here, we report that ZAKα and NLRP1 sense bacterial exotoxins that target ribosome elongation factors. One such toxin, diphtheria toxin (DT), the causative agent for human diphtheria, triggers RSR-dependent inflammasome activation in primary human keratinocytes. This process requires iron-mediated DT production in the bacteria, as well as diphthamide synthesis and ZAKα/p38-driven NLRP1 phosphorylation in host cells. NLRP1 deletion abrogates IL-1β and IL-18 secretion by DT-intoxicated keratinocytes, while ZAKα deletion or inhibition additionally limits both pyroptotic and inflammasome-independent non-pyroptotic cell death. Consequently, pharmacologic inhibition of ZAKα is more effective than caspase-1 inhibition at protecting the epidermal barrier in a 3D skin model of cutaneous diphtheria. In summary, these findings implicate ZAKα-driven RSR and the NLRP1 inflammasome in antibacterial immunity and might explain certain aspects of diphtheria pathogenesis.
Collapse
Affiliation(s)
- Kim Samirah Robinson
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- The A*STAR Skin Research Labs, Singapore, Singapore
| | - Gee Ann Toh
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | | | | | - Pritisha Rozario
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Chrissie K. Lim
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| | - Ying Xiu Toh
- The A*STAR Skin Research Labs, Singapore, Singapore
| | - Zhi Heng Lau
- The A*STAR Skin Research Labs, Singapore, Singapore
| | | | - Jacob Mayer
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | | | - Florian I. Schmidt
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | | | - Franklin L. Zhong
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Skin Research Institute of Singapore, Singapore, Singapore
| |
Collapse
|
17
|
Tatsukawa T, Kano K, Nakajima KI, Yazawa T, Eguchi R, Kabara M, Horiuchi K, Hayasaka T, Matsuo R, Hasebe N, Azuma N, Kawabe JI. NG2-positive pericytes regulate homeostatic maintenance of slow-type skeletal muscle with rapid myonuclear turnover. Stem Cell Res Ther 2023; 14:205. [PMID: 37592340 PMCID: PMC10433572 DOI: 10.1186/s13287-023-03433-1] [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: 01/24/2023] [Accepted: 07/26/2023] [Indexed: 08/19/2023] Open
Abstract
BACKGROUND Skeletal muscle comprises almost 40% of the human body and is essential for movement, structural support and metabolic homeostasis. Size of multinuclear skeletal muscle is stably maintained under steady conditions with the sporadic fusion of newly produced myocytes to compensate for the muscular turnover caused by daily wear and tear. It is becoming clear that microvascular pericytes (PCs) exhibit myogenic activity. However, whether PCs act as myogenic stem cells for the homeostatic maintenance of skeletal muscles during adulthood remains uncertain. METHODS We utilized PC-fused myofibers using PC-specific lineage tracing mouse (NG2-CreERT/Rosa-tdTomato) to observe whether muscle resident PCs have myogenic potential during daily life. Genetic PC deletion mouse model (NG2-CreERT/DTA) was used to test whether PC differentiates to myofibers for maintenance of muscle structure and function under homeostatic condition. RESULTS Under steady breeding conditions, tdTomato-expressing PCs were infused into myofibers, and subsequently, PC-derived nuclei were incorporated into myofibers. Especially in type-I slow-type myofibers such as the soleus, tdTomato+ myofibers were already observed 3 days after PC labeling; their ratio reached a peak (approximately 80%) within 1 month and was maintained for more than 1 year. Consistently, the NG2+ PC-specific deletion induced muscular atrophy in a slow-type myofiber-specific manner under steady breeding conditions. The number of myonucleus per volume of each myofiber was constant during observation period. CONCLUSIONS These findings demonstrate that the turnover of myonuclei in slow-type myofibers is relatively fast, with PCs acting as myogenic stem cells-the suppliers of new myonuclei under steady conditions-and play a vital role in the homeostatic maintenance of slow-type muscles.
Collapse
Affiliation(s)
- Takamitsu Tatsukawa
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
- Department of Vascular Surgery, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Kohei Kano
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
- Department of Cardiovascular Regeneration and Innovation, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Kei-Ichi Nakajima
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Takashi Yazawa
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Ryoji Eguchi
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Maki Kabara
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Kiwamu Horiuchi
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
- Division of Cardiovascular, Respiratory and Neurology, Department of Medicine, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Taiki Hayasaka
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
- Division of Cardiovascular, Respiratory and Neurology, Department of Medicine, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Risa Matsuo
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
- Department of Dermatology, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Naoyuki Hasebe
- Department of Cardiovascular Regeneration and Innovation, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
- Division of Cardiovascular, Respiratory and Neurology, Department of Medicine, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Nobuyoshi Azuma
- Department of Vascular Surgery, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Jun-Ichi Kawabe
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan.
- Department of Cardiovascular Regeneration and Innovation, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan.
| |
Collapse
|
18
|
Deng Y, He Y, Xu J, He H, Li G. Heterogeneity and Functional Analysis of Cardiac Fibroblasts in Heart Development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.30.551164. [PMID: 37577541 PMCID: PMC10418062 DOI: 10.1101/2023.07.30.551164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Background As one of the major cell types in the heart, fibroblasts play critical roles in multiple biological processes. Cardiac fibroblasts are known to develop from multiple sources, but their transcriptional profiles have not been systematically compared. Furthermore, while the function of a few genes in cardiac fibroblasts has been studied, the overall function of fibroblasts as a cell type remains uninvestigated. Methods Single-cell mRNA sequencing (scRNA-seq) and bioinformatics approaches were used to analyze the genome-wide genes expression and extracellular matrix genes expression in fibroblasts, as well as the ligand-receptor interactions between fibroblasts and cardiomyocytes. Single molecular in situ hybridization was employed to analyze the expression pattern of fibroblast subpopulation-specific genes. The Diphtheria toxin fragment A (DTA) system was utilized to ablate fibroblasts at each developmental phase. Results Using RNA staining of Col1a1 at different stages, we grouped cardiac fibroblasts into four developmental phases. Through the analysis of scRNA-seq profiles of fibroblasts at 18 stages from two mouse strains, we identified significant heterogeneity, preserving lineage gene expression in their precursor cells. Within the main fibroblast population, we found differential expressions of Wt1, Tbx18, and Aldh1a2 genes in various cell clusters. Lineage tracing studies showed Wt1- and Tbx18-positive fibroblasts originated from respective epicardial cells. Furthermore, using a conditional DTA system-based elimination, we identified the crucial role of fibroblasts in early embryonic and heart growth, but not in neonatal heart growth. Additionally, we identified the zone- and stage-associated expression of extracellular matrix genes and fibroblast-cardiomyocyte ligand-receptor interactions. This comprehensive understanding sheds light on fibroblast function in heart development. Conclusion We observed cardiac fibroblast heterogeneity at embryonic and neonatal stages, with preserved lineage gene expression. Ablation studies revealed their distinct roles during development, likely influenced by varying extracellular matrix genes and ligand-receptor interactions at different stages.
Collapse
|
19
|
Liao Y, Xiang Y, Zheng M, Wang J. DeepMiceTL: a deep transfer learning based prediction of mice cardiac conduction diseases using early electrocardiograms. Brief Bioinform 2023; 24:bbad109. [PMID: 36935112 PMCID: PMC10422927 DOI: 10.1093/bib/bbad109] [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: 11/29/2022] [Revised: 02/10/2023] [Accepted: 03/01/2023] [Indexed: 03/21/2023] Open
Abstract
Cardiac conduction disease is a major cause of morbidity and mortality worldwide. There is considerable clinical significance and an emerging need of early detection of these diseases for preventive treatment success before more severe arrhythmias occur. However, developing such early screening tools is challenging due to the lack of early electrocardiograms (ECGs) before symptoms occur in patients. Mouse models are widely used in cardiac arrhythmia research. The goal of this paper is to develop deep learning models to predict cardiac conduction diseases in mice using their early ECGs. We hypothesize that mutant mice present subtle abnormalities in their early ECGs before severe arrhythmias present. These subtle patterns can be detected by deep learning though they are hard to be identified by human eyes. We propose a deep transfer learning model, DeepMiceTL, which leverages knowledge from human ECGs to learn mouse ECG patterns. We further apply the Bayesian optimization and $k$-fold cross validation methods to tune the hyperparameters of the DeepMiceTL. Our results show that DeepMiceTL achieves a promising performance (F1-score: 83.8%, accuracy: 84.8%) in predicting the occurrence of cardiac conduction diseases using early mouse ECGs. This study is among the first efforts that use state-of-the-art deep transfer learning to identify ECG patterns during the early course of cardiac conduction disease in mice. Our approach not only could help in cardiac conduction disease research in mice, but also suggest a feasibility for early clinical diagnosis of human cardiac conduction diseases and other types of cardiac arrythmias using deep transfer learning in the future.
Collapse
Affiliation(s)
- Ying Liao
- Department of Industrial, Manufacturing & Systems Engineering, Texas Tech University, Lubbock, Texas, USA
| | - Yisha Xiang
- Department of Industrial Engineering, University of Houston, Houston, Texas, USA
| | - Mingjie Zheng
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Jun Wang
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
20
|
Cronin SJF, Tejada MA, Song R, Laval K, Cikes D, Ji M, Brai A, Stadlmann J, Novatchikova M, Perlot T, Ali OH, Botta L, Decker T, Lazovic J, Hagelkruys A, Enquist L, Rao S, Koyuncu OO, Penninger JM. Pseudorabies virus hijacks DDX3X, initiating an addictive "mad itch" and immune suppression, to facilitate viral spread. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.09.539956. [PMID: 37214906 PMCID: PMC10197578 DOI: 10.1101/2023.05.09.539956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Infections with defined Herpesviruses, such as Pseudorabies virus (PRV) and Varicella zoster virus (VZV) can cause neuropathic itch, referred to as "mad itch" in multiple species. The underlying mechanisms involved in neuropathic "mad itch" are poorly understood. Here, we show that PRV infections hijack the RNA helicase DDX3X in sensory neurons to facilitate anterograde transport of the virus along axons. PRV induces re-localization of DDX3X from the cell body to the axons which ultimately leads to death of the infected sensory neurons. Inducible genetic ablation of Ddx3x in sensory neurons results in neuronal death and "mad itch" in mice. This neuropathic "mad itch" is propagated through activation of the opioid system making the animals "addicted to itch". Moreover, we show that PRV co-opts and diverts T cell development in the thymus via a sensory neuron-IL-6-hypothalamus-corticosterone stress pathway. Our data reveal how PRV, through regulation of DDX3X in sensory neurons, travels along axons and triggers neuropathic itch and immune deviations to initiate pathophysiological programs which facilitate its spread to enhance infectivity.
Collapse
Affiliation(s)
- Shane J F Cronin
- Institute of Molecular Biotechnology Austria (IMBA), Dr. Bohrgasse 3, A-1030 Vienna, Austria
| | - Miguel A Tejada
- Institute of Molecular Biotechnology Austria (IMBA), Dr. Bohrgasse 3, A-1030 Vienna, Austria
| | - Ren Song
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | - Kathlyn Laval
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
- Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Domagoj Cikes
- Institute of Molecular Biotechnology Austria (IMBA), Dr. Bohrgasse 3, A-1030 Vienna, Austria
| | - Ming Ji
- Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Annalaura Brai
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, I-53100 Siena, Italy
| | - Johannes Stadlmann
- Institute of Molecular Biotechnology Austria (IMBA), Dr. Bohrgasse 3, A-1030 Vienna, Austria
| | - Maria Novatchikova
- Institute of Molecular Biotechnology Austria (IMBA), Dr. Bohrgasse 3, A-1030 Vienna, Austria
| | - Thomas Perlot
- Institute of Molecular Biotechnology Austria (IMBA), Dr. Bohrgasse 3, A-1030 Vienna, Austria
| | - Omar Hasan Ali
- Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, Canada
- Institute of Immunobiology, Cantonal Hospital St. Gallen, Rorschacher Strasse 95, 9007 St. Gallen, Switzerland
- Department of Dermatology, University of Zurich, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Lorenzo Botta
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, I-53100 Siena, Italy
| | - Thomas Decker
- Max F. Perutz Laboratories, Department of Microbiology, Immunobiology and Genetics, University of Vienna, Vienna Biocenter, Vienna, Austria
| | - Jelena Lazovic
- Institute of Molecular Biotechnology Austria (IMBA), Dr. Bohrgasse 3, A-1030 Vienna, Austria
| | - Astrid Hagelkruys
- Institute of Molecular Biotechnology Austria (IMBA), Dr. Bohrgasse 3, A-1030 Vienna, Austria
| | - Lynn Enquist
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | - Shuan Rao
- Department of Thoracic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Orkide O Koyuncu
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
- Department of Microbiology and Molecular Genetics, School of Medicine, University of California, Irvine, Irvine, CA 92697-4025, USA
| | - Josef M Penninger
- Institute of Molecular Biotechnology Austria (IMBA), Dr. Bohrgasse 3, A-1030 Vienna, Austria
- Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| |
Collapse
|
21
|
Goodwin K, Nelson CM. Analysis of Cre lines for targeting embryonic airway smooth muscle. Dev Biol 2023; 496:63-72. [PMID: 36706974 PMCID: PMC10041960 DOI: 10.1016/j.ydbio.2023.01.008] [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: 08/04/2022] [Revised: 01/09/2023] [Accepted: 01/22/2023] [Indexed: 01/26/2023]
Abstract
During development of the embryonic mouse lung, the pulmonary mesenchyme differentiates into smooth muscle that wraps around the airway epithelium. Inhibiting smooth muscle differentiation leads to cystic airways, while enhancing it stunts epithelial branching. These findings support a conceptual model wherein the differentiation of smooth muscle sculpts the growing epithelium into branches at precise positions and with stereotyped morphologies. Unfortunately, most approaches to manipulate the differentiation of airway smooth muscle rely on pharmacological or physical perturbations that are conducted ex vivo. Here, we explored the use of diphtheria toxin-based genetic ablation strategies to eliminate airway smooth muscle in the embryonic mouse lung. Surprisingly, neither airway smooth muscle wrapping nor epithelial branching were affected in embryos in which the expression of diphtheria toxin or its receptor were driven by several different smooth muscle-specific Cre lines. Close examination of spatial patterns of Cre activity in the embryonic lung revealed that none of these commonly used Cre lines target embryonic airway smooth muscle robustly or specifically. Our findings demonstrate the need for airway smooth muscle-specific Cre lines that are active in the embryonic lung, and serve as a resource for researchers contemplating the use of these commonly used Cre lines for studying embryonic airway smooth muscle.
Collapse
Affiliation(s)
- Katharine Goodwin
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, 08544, USA
| | - Celeste M Nelson
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, 08544, USA; Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA.
| |
Collapse
|
22
|
Meller SJ, Hernandez L, Martin-Lopez E, Kloos ZA, Liberia T, Greer CA. Microglia Maintain Homeostatic Conditions in the Developing Rostral Migratory Stream. eNeuro 2023; 10:ENEURO.0197-22.2023. [PMID: 36697258 PMCID: PMC9910579 DOI: 10.1523/eneuro.0197-22.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 01/03/2023] [Accepted: 01/14/2023] [Indexed: 01/27/2023] Open
Abstract
Microglia invade the neuroblast migratory corridor of the rostral migratory stream (RMS) early in development. The early postnatal RMS does not yet have the dense astrocyte and vascular scaffold that helps propel forward migrating neuroblasts, which led us to consider whether microglia help regulate conditions permissive to neuroblast migration in the RMS. GFP-labeled microglia in CX3CR-1GFP/+ mice assemble primarily along the outer borders of the RMS during the first postnatal week, where they exhibit predominantly an ameboid morphology and associate with migrating neuroblasts. Microglia ablation for 3 d postnatally does not impact the density of pulse labeled BrdU+ neuroblasts nor the distance migrated by tdTomato electroporated neuroblasts in the RMS. However, microglia wrap DsRed-labeled neuroblasts in the RMS of P7 CX3CR-1GFP/+;DCXDsRed/+ mice and express the markers CD68, CLEC7A, MERTK, and IGF-1, suggesting active regulation in the developing RMS. Microglia depletion for 14 d postnatally further induced an accumulation of CC3+ DCX+ apoptotic neuroblasts in the RMS, a wider RMS and extended patency of the lateral ventricle extension in the olfactory bulb. These findings illustrate the importance of microglia in maintaining a healthy neuroblast population and an environment permissive to neuroblast migration in the early postnatal RMS.
Collapse
Affiliation(s)
- Sarah J Meller
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06520
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT 06520
- The Interdepartmental Neuroscience Graduate Program, Yale University School of Medicine, New Haven, CT 06520
| | - Lexie Hernandez
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06520
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT 06520
| | - Eduardo Martin-Lopez
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06520
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT 06520
| | - Zachary A Kloos
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06520
| | - Teresa Liberia
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06520
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT 06520
| | - Charles A Greer
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06520
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT 06520
- The Interdepartmental Neuroscience Graduate Program, Yale University School of Medicine, New Haven, CT 06520
| |
Collapse
|
23
|
Jia S, Wilbourne J, Crossen MJ, Zhao F. Morphogenesis of the female reproductive tract along antero-posterior and dorso-ventral axes is dependent on Amhr2+ mesenchyme in mice†. Biol Reprod 2022; 107:1477-1489. [PMID: 36130202 PMCID: PMC9752753 DOI: 10.1093/biolre/ioac179] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 08/11/2022] [Accepted: 09/15/2022] [Indexed: 12/24/2022] Open
Abstract
Morphogenesis of the female reproductive tract is regulated by the mesenchyme. However, the identity of the mesenchymal lineage that directs the morphogenesis of the female reproductive tract has not been determined. Using in vivo genetic cell ablation, we identified Amhr2+ mesenchyme as an essential mesenchymal population in patterning the female reproductive tract. After partial ablation of Amhr2+ mesenchymal cells, the oviduct failed to develop its characteristic coiling due to decreased epithelial proliferation and tubule elongation during development. The uterus displayed a reduction in size and showed decreased cellular proliferation in both epithelial and mesenchymal compartments. More importantly, in the uterus, partial ablation of Amhr2+ mesenchyme caused abnormal lumen shape and altered the direction of its long axis from the dorsal-ventral axis to the left-right axis (i.e., perpendicular to the dorsal-ventral axis). Despite these morphological defects, epithelia underwent normal differentiation into secretory and ciliated cells in the oviduct and glandular epithelial cells in the uterus. These results demonstrated that Amhr2+ mesenchyme can direct female reproductive tract morphogenesis by regulating epithelial proliferation and lumen shape without affecting the differentiation of epithelial cell types.
Collapse
Affiliation(s)
- Shuai Jia
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Jillian Wilbourne
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - McKenna J Crossen
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
- Endocrinology and Reproductive Physiology Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Fei Zhao
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
- Endocrinology and Reproductive Physiology Program, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
24
|
Konkimalla A, Konishi S, Kobayashi Y, Kadur Lakshminarasimha Murthy P, Macadlo L, Mukherjee A, Elmore Z, Kim SJ, Pendergast AM, Lee PJ, Asokan A, Knudsen L, Bravo-Cordero JJ, Tata A, Tata PR. Multi-apical polarity of alveolar stem cells and their dynamics during lung development and regeneration. iScience 2022; 25:105114. [PMID: 36185377 PMCID: PMC9519774 DOI: 10.1016/j.isci.2022.105114] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 08/25/2022] [Accepted: 09/08/2022] [Indexed: 11/24/2022] Open
Abstract
Epithelial cells of diverse tissues are characterized by the presence of a single apical domain. In the lung, electron microscopy studies have suggested that alveolar type-2 epithelial cells (AT2s) en face multiple alveolar sacs. However, apical and basolateral organization of the AT2s and their establishment during development and remodeling after injury repair remain unknown. Thick tissue imaging and electron microscopy revealed that a single AT2 can have multiple apical domains that enface multiple alveoli. AT2s gradually establish multi-apical domains post-natally, and they are maintained throughout life. Lineage tracing, live imaging, and selective cell ablation revealed that AT2s dynamically reorganize multi-apical domains during injury repair. Single-cell transcriptome signatures of residual AT2s revealed changes in cytoskeleton and cell migration. Significantly, cigarette smoke and oncogene activation lead to dysregulation of multi-apical domains. We propose that the multi-apical domains of AT2s enable them to be poised to support the regeneration of a large array of alveolar sacs.
Collapse
Affiliation(s)
- Arvind Konkimalla
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
- Medical Scientist Training Program, Duke University School of Medicine, Durham, NC 27710, USA
| | - Satoshi Konishi
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Yoshihiko Kobayashi
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | | | - Lauren Macadlo
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Ananya Mukherjee
- Division of Hematology and Oncology, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Zachary Elmore
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - So-Jin Kim
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Duke University School of Medicine and the Durham Veterans Administration Medical Center, Durham, NC 27710, USA
| | - Ann Marie Pendergast
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710, USA
| | - Patty J. Lee
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Duke University School of Medicine and the Durham Veterans Administration Medical Center, Durham, NC 27710, USA
| | - Aravind Asokan
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Biomedical Engineering, Regeneration Next, Duke University, Durham, NC 27710, USA
- Center for Advanced Genomic Technologies, Duke University, Durham, NC 27710, USA
| | - Lars Knudsen
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover 30625, Germany
| | - Jose Javier Bravo-Cordero
- Division of Hematology and Oncology, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Aleksandra Tata
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Purushothama Rao Tata
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Duke University School of Medicine and the Durham Veterans Administration Medical Center, Durham, NC 27710, USA
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC 27710, USA
- Center for Advanced Genomic Technologies, Duke University, Durham, NC 27710, USA
- Duke Regeneration Center, Duke University, Durham, NC 27710, USA
| |
Collapse
|
25
|
Schmidt M, Sondermann JR, Gomez-Varela D, Çubuk C, Millet Q, Lewis MJ, Wood JN, Zhao J. Transcriptomic and proteomic profiling of Na V1.8-expressing mouse nociceptors. Front Mol Neurosci 2022; 15:1002842. [PMID: 36305001 PMCID: PMC9593034 DOI: 10.3389/fnmol.2022.1002842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 09/14/2022] [Indexed: 11/18/2022] Open
Affiliation(s)
- Manuela Schmidt
- Systems Biology of Pain, Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| | - Julia Regina Sondermann
- Systems Biology of Pain, Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| | - David Gomez-Varela
- Systems Biology of Pain, Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| | - Cankut Çubuk
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Centre for Translational Bioinformatics, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Queensta Millet
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, Division of Medicine, University College London, London, United Kingdom
| | - Myles J. Lewis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
- Centre for Translational Bioinformatics, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - John N. Wood
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, Division of Medicine, University College London, London, United Kingdom
| | - Jing Zhao
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, Division of Medicine, University College London, London, United Kingdom
| |
Collapse
|
26
|
Li SY, Bhandary B, Gu X, DeFalco T. Perivascular cells support folliculogenesis in the developing ovary. Proc Natl Acad Sci U S A 2022; 119:e2213026119. [PMID: 36194632 PMCID: PMC9564831 DOI: 10.1073/pnas.2213026119] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 09/07/2022] [Indexed: 11/18/2022] Open
Abstract
Supporting cells of the ovary, termed granulosa cells, are essential for ovarian differentiation and oogenesis by providing a nurturing environment for oocyte maintenance and maturation. Granulosa cells are specified in the fetal and perinatal ovary, and sufficient numbers of granulosa cells are critical for the establishment of follicles and the oocyte reserve. Identifying the cellular source from which granulosa cells and their progenitors are derived is an integral part of efforts to understand basic ovarian biology and the etiology of female infertility. In particular, the contribution of mesenchymal cells, especially perivascular cells, to ovarian development is poorly understood but is likely to be a source of new information regarding ovarian function. Here we have identified a cell population in the fetal ovary, which is a Nestin-expressing perivascular cell type. Using lineage tracing and ex vivo organ culture methods, we determined that perivascular cells are multipotent progenitors that contribute to granulosa, thecal, and pericyte cell lineages in the ovary. Maintenance of these progenitors is dependent on ovarian vasculature, likely reliant on endothelial-mesenchymal Notch signaling interactions. Depletion of Nestin+ progenitors resulted in a disruption of granulosa cell specification and in an increased number of germ cell cysts that fail to break down, leading to polyovular ovarian follicles. These findings highlight a cell population in the ovary and uncover a key role for vasculature in ovarian differentiation, which may lead to insights into the origins of female gonad dysgenesis and infertility.
Collapse
Affiliation(s)
- Shu-Yun Li
- Reproductive Sciences Center, Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229
| | - Bidur Bhandary
- Reproductive Sciences Center, Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229
| | - Xiaowei Gu
- Reproductive Sciences Center, Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229
| | - Tony DeFalco
- Reproductive Sciences Center, Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267
| |
Collapse
|
27
|
Sartiani L, Bartolucci G, Pallecchi M, Spinelli V, Cerbai E. Pharmacological basis of the antifibrotic effects of pirfenidone: Mechanistic insights from cardiac in-vitro and in-vivo models. Front Cardiovasc Med 2022; 9:751499. [PMID: 36204580 PMCID: PMC9530045 DOI: 10.3389/fcvm.2022.751499] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 08/19/2022] [Indexed: 02/06/2023] Open
Abstract
Pirfenidone is a small drug with marked antifibrotic activity approved for the treatment of Idiopathic pulmonary fibrosis. Recently, its peculiar pharmacological profile has attracted attention for its potential therapeutic benefit for extra-pulmonary disorders characterized by pathological fibrosis, such as kidney, liver, and cardiac failure. A major pitfall of pirfenidone is the lack of consistent understanding of its mechanism of action, regardless of the target. In addition to the increasing attention to the role of inflammation and its mediators in several processes, a better knowledge of the variety of fibroblasts' population, of signals controlling their activation and trans-differentiation, and of crosstalk with other cell resident and non-resident cell types is needed for prevention, treatment and possibly reverse of fibrosis. This review will focus on pirfenidone's pharmacological profile and its effects on cardiac fibroblasts.
Collapse
Affiliation(s)
- Laura Sartiani
- Section of Pharmacology, Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Gianluca Bartolucci
- Section of Medicinal Chemistry, Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Marco Pallecchi
- Section of Medicinal Chemistry, Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Valentina Spinelli
- Section of Pharmacology, Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Elisabetta Cerbai
- Section of Pharmacology, Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy
- *Correspondence: Elisabetta Cerbai
| |
Collapse
|
28
|
Hwang SJ, Drumm BT, Kim MK, Lyu JH, Baker S, Sanders KM, Ward SM. Calcium transients in intramuscular interstitial cells of Cajal of the murine gastric fundus and their regulation by neuroeffector transmission. J Physiol 2022; 600:4439-4463. [PMID: 36057845 DOI: 10.1113/jp282876] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 08/15/2022] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS The cells responsible for mediating enteric neuroeffector transmission remain controversial. In the stomach intramuscular interstitial cells of Cajal (ICC-IM) were the first ICC reported to receive cholinergic and nitrergic neural inputs. Utilization of a cell specific calcium biosensor, GCaMP6f, the activity and neuroeffector responses of ICC-IM were examined. ICC-IM were highly active, generating stochastic intracellular Ca2+ -transients. Stimulation of enteric motor nerves abolished Ca2+ -transients in ICC-IM. This inhibitory response was preceded by a global rise in intracellular Ca2+ . Individual ICC-IM responded to nerve stimulation with a rise in Ca2+ followed by inhibition of Ca2+ -transients. Inhibition of Ca2+ -transients was blocked by the nitric oxide synthase antagonist, L-NNA. The global rise in Ca2+ was inhibited by the muscarinic antagonist, atropine. Simultaneous intracellular recordings with video imaging revealed that the global rise in intracellular Ca2+ and inhibition of Ca2+ -transients was temporally associated with rapid excitatory junction potentials followed by more sustained inhibitory junction potentials. The data presented support the premise of serial innervation of ICC-IM in excitatory and inhibitory neuroeffector transmission in the proximal stomach. ABSTRACT Enteric neurotransmission is critical for coordinating motility throughout the gastrointestinal (GI) tract. However, there is considerable controversy regarding the cells that are responsible for the transduction of these neural inputs. In the present study, utilization of a cell-specific calcium biosensor GCaMP6f, the spontaneous activity and neuroeffector responses of intramuscular ICC (ICC-IM) to motor neural inputs was examined. Simultaneous intracellular microelectrode recordings and high-speed video-imaging during nerve stimulation was used to reveal the temporal relationship between changes in intracellular Ca2+ and post-junctional electrical responses to neural stimulation. ICC-IM were highly active, generating intracellular Ca2+ -transients that occurred stochastically, from multiple independent sites in single ICC-IM. Ca2+ -transients were not entrained in single ICC-IM or between neighboring ICC-IM. Activation of enteric motor neurons produced a dominant inhibitory response that abolished Ca2+ -transients in ICC-IM. This inhibitory response was often preceded by a summation of Ca2+ -transients that led to a global rise in Ca2+ . Individual ICC-IM responded to nerve stimulation by a global rise in Ca2+ followed by inhibition of Ca2+ -transients. The inhibition of Ca2+ -transients was blocked by the nitric oxide synthase antagonist, L-NNA. The global rise in intracellular Ca2+ was inhibited by the muscarinic antagonist, atropine. Simultaneous intracellular microelectrode recordings with video-imaging revealed that the rise in Ca2+ was temporally associated with rapid excitatory junction potentials and the inhibition of Ca2+ -transients with inhibitory junction potentials. These data support the premise of serial innervation of ICC-IM in excitatory and inhibitory neuroeffector transmission in the proximal stomach. Abstract figure legend Intramuscular interstitial cells of Cajal (ICC-IM) of the gastric fundus receive nitrergic inhibitory and cholinergic excitatory neuroeffector motor inputs. Using a genetically encoded calcium sensor we demonstrate that ICC-IM are highly active cells generating stochastic intracellular Ca2 -transients. Stimulation of enteric motor nerves abolished Ca2 -transients in ICC-IM, produced an inhibitory junction potential (IJP) and muscle relaxation that was mediated by nitric oxide (left hand side of figure). This inhibitory response was often preceded by a global rise in intracellular Ca2 in ICC-IM, a rapid excitatory junction potential (EJP) and muscle contraction, that was mediated by acetylcholine (right hand side of figure). Individual ICC-IM could respond to both excitatory and inhibitory neural inputs. These data support the premise of serial innervation of ICC-IM in excitatory and inhibitory neuroeffector transmission in the proximal stomach. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Sung Jin Hwang
- Department of Physiology & Cell Biology, Reno School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Bernard T Drumm
- Department of Physiology & Cell Biology, Reno School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Min Kyung Kim
- Department of Physiology & Cell Biology, Reno School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Ju Hyeong Lyu
- Department of Physiology & Cell Biology, Reno School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Sal Baker
- Department of Physiology & Cell Biology, Reno School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Kenton M Sanders
- Department of Physiology & Cell Biology, Reno School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Sean M Ward
- Department of Physiology & Cell Biology, Reno School of Medicine, University of Nevada, Reno, NV, 89557, USA
| |
Collapse
|
29
|
Zhao F, Grimm SA, Jia S, Yao HHC. Contribution of the Wolffian duct mesenchyme to the formation of the female reproductive tract. PNAS NEXUS 2022; 1:pgac182. [PMID: 36204418 PMCID: PMC9523451 DOI: 10.1093/pnasnexus/pgac182] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 09/06/2022] [Indexed: 02/05/2023]
Abstract
The female reproductive tract develops from its embryonic precursor, the Müllerian duct. In close proximity to the Müllerian duct lies the precursor for the male reproductive tract, the Wolffian duct, which is eliminated in the female embryo during sexual differentiation. We discovered that a component of the Wolffian duct, its mesenchyme, is not eliminated after sexual differentiation. Instead, the Wolffian duct mesenchyme underwent changes in transcriptome and chromatin accessibility from male tract to female tract identity, and became a unique mesenchymal population in the female reproductive tract with localization and transcriptome distinct from the mesenchyme derived from the Müllerian duct. Partial ablation of the Wolffian duct mesenchyme stunted the growth of the fetal female reproductive tract in ex vivo organ culture. These findings reveal a new fetal origin of mesenchymal tissues for female reproductive tract formation and reshape our understanding of sexual differentiation of reproductive tracts.
Collapse
Affiliation(s)
- Fei Zhao
- Reproductive Developmental Biology Group, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Sara A Grimm
- Integrative Bioinformatics Support Group, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Shua Jia
- Present address: Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin–Madison, Madison, WI 53706, USA
| | - Humphrey Hung-Chang Yao
- Reproductive Developmental Biology Group, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| |
Collapse
|
30
|
Mapps AA, Boehm E, Beier C, Keenan WT, Langel J, Liu M, Thomsen MB, Hattar S, Zhao H, Tampakakis E, Kuruvilla R. Satellite glia modulate sympathetic neuron survival, activity, and autonomic function. eLife 2022; 11:74295. [PMID: 35997251 PMCID: PMC9433091 DOI: 10.7554/elife.74295] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
Satellite glia are the major glial cells in sympathetic ganglia, enveloping neuronal cell bodies. Despite this intimate association, the extent to which sympathetic functions are influenced by satellite glia in vivo remains unclear. Here, we show that satellite glia are critical for metabolism, survival, and activity of sympathetic neurons and modulate autonomic behaviors in mice. Adult ablation of satellite glia results in impaired mTOR signaling, soma atrophy, reduced noradrenergic enzymes, and loss of sympathetic neurons. However, persisting neurons have elevated activity, and satellite glia-ablated mice show increased pupil dilation and heart rate, indicative of enhanced sympathetic tone. Satellite glia-specific deletion of Kir4.1, an inward-rectifying potassium channel, largely recapitulates the cellular defects observed in glia-ablated mice, suggesting that satellite glia act in part via K+-dependent mechanisms. These findings highlight neuron–satellite glia as functional units in regulating sympathetic output, with implications for disorders linked to sympathetic hyper-activity such as cardiovascular disease and hypertension.
Collapse
Affiliation(s)
- Aurelia A Mapps
- Department of Biology, Johns Hopkins University, Baltimore, United States
| | - Erica Boehm
- Department of Biology, Johns Hopkins University, Baltimore, United States
| | - Corinne Beier
- Section on Light and Circadian Rhythms (SLCR), National Institute of Mental Health, Bethesda, United States
| | - William T Keenan
- Department of Biology, Johns Hopkins University, Baltimore, United States
| | - Jennifer Langel
- Section on Light and Circadian Rhythms (SLCR), National Institute of Mental Health, Bethesda, United States
| | - Michael Liu
- Department of Biology, Johns Hopkins University, Baltimore, United States
| | - Michael B Thomsen
- Section on Light and Circadian Rhythms (SLCR), National Institute of Mental Health, Bethesda, United States
| | - Samer Hattar
- Section on Light and Circadian Rhythms (SLCR), National Institute of Mental Health, Bethesda, United States
| | - Haiqing Zhao
- Department of Biology, Johns Hopkins University, Baltimore, United States
| | | | - Rejji Kuruvilla
- Department of Biology, Johns Hopkins University, Baltimore, United States
| |
Collapse
|
31
|
Steele RE, Sanders R, Phillips HM, Bamforth SD. PAX Genes in Cardiovascular Development. Int J Mol Sci 2022; 23:7713. [PMID: 35887061 PMCID: PMC9324344 DOI: 10.3390/ijms23147713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/07/2022] [Accepted: 07/11/2022] [Indexed: 01/25/2023] Open
Abstract
The mammalian heart is a four-chambered organ with systemic and pulmonary circulations to deliver oxygenated blood to the body, and a tightly regulated genetic network exists to shape normal development of the heart and its associated major arteries. A key process during cardiovascular morphogenesis is the septation of the outflow tract which initially forms as a single vessel before separating into the aorta and pulmonary trunk. The outflow tract connects to the aortic arch arteries which are derived from the pharyngeal arch arteries. Congenital heart defects are a major cause of death and morbidity and are frequently associated with a failure to deliver oxygenated blood to the body. The Pax transcription factor family is characterised through their highly conserved paired box and DNA binding domains and are crucial in organogenesis, regulating the development of a wide range of cells, organs and tissues including the cardiovascular system. Studies altering the expression of these genes in murine models, notably Pax3 and Pax9, have found a range of cardiovascular patterning abnormalities such as interruption of the aortic arch and common arterial trunk. This suggests that these Pax genes play a crucial role in the regulatory networks governing cardiovascular development.
Collapse
Affiliation(s)
| | | | | | - Simon D. Bamforth
- Bioscience Institute, Faculty of Medical Sciences, Newcastle University, Centre for Life, Newcastle NE1 3BZ, UK; (R.E.S.); (R.S.); (H.M.P.)
| |
Collapse
|
32
|
Tseng PY, Hoon MA. GPR15L is an epithelial inflammation-derived pruritogen. SCIENCE ADVANCES 2022; 8:eabm7342. [PMID: 35704588 PMCID: PMC9200282 DOI: 10.1126/sciadv.abm7342] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 05/02/2022] [Indexed: 05/09/2023]
Abstract
Itch is an unpleasant sensation that often accompanies chronic dermatological conditions. Although many of the itch receptors and the neural pathways underlying this sensation are known, the identity of endogenous ligands is still not fully appreciated. Using an unbiased bioinformatic approach, we identified GPR15L as a candidate pruritogen whose expression is robustly up-regulated in psoriasis and atopic dermatitis. Although GPR15L was previously shown to be a cognate ligand of the receptor GPR15, expressed in dermal T cells, here we show that it also contributes to pruritogenesis by activating Mas-related G protein-coupled receptors (MRGPRs). GPR15L can selectively stimulate mouse dorsal root ganglion neurons that express Mrgpra3 and evokes intense itch responses. GPR15L causes mast cell degranulation through stimulation of MRGPRX2 and Mrgprb2. Genetic disruption of GPR15L expression attenuates scratch responses in a mouse model of psoriasis. Our study reveals unrecognized features of GRP15L, showing that it is a potent itch-inducing agent.
Collapse
Affiliation(s)
- Pang-Yen Tseng
- Molecular Genetics Section, National Institute of Dental and Craniofacial Research/NIH, 35 Convent Drive, Bethesda, MD 20892, USA
| | | |
Collapse
|
33
|
Wang K, Xu C, Xie X, Jing Y, Chen P, Yadav S, Wang Z, Taylor R, Wang J, Feng J. Axin2+ PDL Cells Directly Contribute to New Alveolar Bone Formation in Response to Orthodontic Tension Force. J Dent Res 2022; 101:695-703. [PMID: 35001706 PMCID: PMC9124907 DOI: 10.1177/00220345211062585] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Wnt-β-catenin signaling plays a key role in orthodontic tooth movement (OTM), a common clinical practice for malocclusion correction. However, its targeted periodontal ligament (PDL) progenitor cells remain largely unclear. In this study, we first showed a synchronized increase in Wnt-β-catenin levels and Axin2+ PDL progenitor cell numbers during OTM using immunostaining of β-catenin in wild-type mice and X-gal staining in the Axin2-LacZ knock-in line. Next, we demonstrated time-dependent increases in Axin2+ PDL progenitors and their progeny cell numbers within PDL and alveolar bones during OTM using a one-time tamoxifen-induced Axin2 tracing line (Axin2CreERT2/+; R26RtdTomato/+). Coimmunostaining images displayed both early and late bone markers (such as RUNX2 and DMP1) in the Axin2Lin PDL cells. Conversely, ablation of Axin2+ PDL cells via one-time tamoxifen-induced diphtheria toxin subunit A (DTA) led to a drastic decrease in osteogenic activity (as reflected by alkaline phosphatase) in PDL and alveolar bone. There was also a decrease in new bone mass and a significant reduction in the mineral apposition rate on both the control side (to a moderate degree) and the OTM side (to a severe degree). Thus, we conclude that the Axin2+ PDL cells (the Wnt-targeted key cells) are highly sensitive to orthodontic tension force and play a critical role in OTM-induced PDL expansion and alveolar bone formation. Future drug development targeting the Axin2+ PDL progenitor cells may accelerate alveolar bone formation during orthodontic treatment.
Collapse
Affiliation(s)
- K. Wang
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, USA
- Division of Orthodontics, University of Connecticut Health Center, Farmington, CT, USA
| | - C. Xu
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, USA
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - X. Xie
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, USA
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Y. Jing
- Department of Orthodontics, Texas A&M University College of Dentistry, Dallas, TX, USA
| | - P.J. Chen
- Division of Orthodontics, University of Connecticut Health Center, Farmington, CT, USA
| | - S. Yadav
- Division of Orthodontics, University of Connecticut Health Center, Farmington, CT, USA
| | - Z. Wang
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, USA
| | - R.W. Taylor
- Department of Orthodontics, Texas A&M University College of Dentistry, Dallas, TX, USA
| | - J. Wang
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, USA
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - J.Q. Feng
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, USA
| |
Collapse
|
34
|
Boyer JG, Huo J, Han S, Havens JR, Prasad V, Lin BL, Kass DA, Song T, Sadayappan S, Khairallah RJ, Ward CW, Molkentin JD. Depletion of skeletal muscle satellite cells attenuates pathology in muscular dystrophy. Nat Commun 2022; 13:2940. [PMID: 35618700 PMCID: PMC9135721 DOI: 10.1038/s41467-022-30619-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 05/03/2022] [Indexed: 11/11/2022] Open
Abstract
Skeletal muscle can repair and regenerate due to resident stem cells known as satellite cells. The muscular dystrophies are progressive muscle wasting diseases underscored by chronic muscle damage that is continually repaired by satellite cell-driven regeneration. Here we generate a genetic strategy to mediate satellite cell ablation in dystrophic mouse models to investigate how satellite cells impact disease trajectory. Unexpectedly, we observe that depletion of satellite cells reduces dystrophic disease features, with improved histopathology, enhanced sarcolemmal stability and augmented muscle performance. Mechanistically, we demonstrate that satellite cells initiate expression of the myogenic transcription factor MyoD, which then induces re-expression of fetal genes in the myofibers that destabilize the sarcolemma. Indeed, MyoD re-expression in wildtype adult skeletal muscle reduces membrane stability and promotes histopathology, while MyoD inhibition in a mouse model of muscular dystrophy improved membrane stability. Taken together these observations suggest that satellite cell activation and the fetal gene program is maladaptive in chronic dystrophic skeletal muscle.
Collapse
Affiliation(s)
- Justin G Boyer
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH, 45229, USA
| | - Jiuzhou Huo
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Sarah Han
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Julian R Havens
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH, 45229, USA
| | - Vikram Prasad
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Brian L Lin
- Division of Cardiology, Johns Hopkins Medical Institutions, Baltimore, MD, 21205, USA
| | - David A Kass
- Division of Cardiology, Johns Hopkins Medical Institutions, Baltimore, MD, 21205, USA
| | - Taejeong Song
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, 45267, USA
| | - Sakthivel Sadayappan
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, 45267, USA
| | | | - Christopher W Ward
- Department of Orthopedics and Center for Biomedical Engineering and Technology (BioMET), University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jeffery D Molkentin
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA.
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH, 45229, USA.
| |
Collapse
|
35
|
Pax3 Hypomorphs Reveal Hidden Pax7 Functional Genetic Compensation in Utero. J Dev Biol 2022; 10:jdb10020019. [PMID: 35645295 PMCID: PMC9149870 DOI: 10.3390/jdb10020019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 04/25/2022] [Accepted: 05/11/2022] [Indexed: 01/14/2023] Open
Abstract
Pax3 and Pax7 transcription factors are paralogs within the Pax gene family that that are expressed in early embryos in partially overlapping expression domains and have distinct functions. Significantly, mammalian development is largely unaffected by Pax7 systemic deletion but systemic Pax3 deletion results in defects in neural tube closure, neural crest emigration, cardiac outflow tract septation, muscle hypoplasia and in utero lethality by E14. However, we previously demonstrated that Pax3 hypomorphs expressing only 20% functional Pax3 protein levels exhibit normal neural tube and heart development, but myogenesis is selectively impaired. To determine why only some Pax3-expressing cell lineages are affected and to further titrate Pax3 threshold levels required for neural tube and heart development, we generated hypomorphs containing both a hypomorphic and a null Pax3 allele. This resulted in mutants only expressing 10% functional Pax3 protein with exacerbated neural tube, neural crest and muscle defects, but still a normal heart. To examine why the cardiac neural crest appears resistant to very low Pax3 levels, we examined its paralog Pax7. Significantly, Pax7 expression is both ectopically expressed in Pax3-expressing dorsal neural tube cells and is also upregulated in the Pax3-expressing lineages. To test whether this compensatory Pax7 expression is functional, we deleted Pax7 both systemically and lineage-specifically in hypomorphs expressing only 10% Pax3. Removal of one Pax7 allele resulted in partial outflow tract defects, and complete loss of Pax7 resulted in full penetrance outflow tract defects and in utero lethality. Moreover, combinatorial loss of Pax3 and Pax7 resulted in severe craniofacial defects and a total block of neural crest cell emigration from the neural tube. Pax7Cre lineage mapping revealed ectopic labeling of Pax3-derived neural crest tissues and within the outflow tract of the heart, experimentally confirming the observation of ectopic activation of Pax7 in 10% Pax3 hypomorphs. Finally, genetic cell ablation of Pax7Cre-marked cells is sufficient to cause outflow tract defects in hypomorphs expressing only 10% Pax3, confirming that ectopic and induced Pax7 can play an overlapping functional genetic compensational role in both cardiac neural crest lineage and during craniofacial development, which is normally masked by the dominant role of Pax3.
Collapse
|
36
|
Qiao L, Saget S, Lu C, Zang T, Dzyuba B, Hay WW, Shao J. The Essential Role of Pancreatic α-Cells in Maternal Metabolic Adaptation to Pregnancy. Diabetes 2022; 71:978-988. [PMID: 35147704 PMCID: PMC9044124 DOI: 10.2337/db21-0923] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 02/07/2022] [Indexed: 11/13/2022]
Abstract
Pancreatic α-cells are important in maintaining metabolic homeostasis, but their role in regulating maternal metabolic adaptations to pregnancy has not been studied. The objective of this study was to determine whether pancreatic α-cells respond to pregnancy and their contribution to maternal metabolic adaptation. With use of C57BL/6 mice, the findings of our study showed that pregnancy induced a significant increase of α-cell mass by promoting α-cell proliferation that was associated with a transitory increase of maternal serum glucagon concentration in early pregnancy. Maternal pancreatic GLP-1 content also was significantly increased during pregnancy. Using the inducible Cre/loxp technique, we ablated the α-cells (α-null) before and during pregnancy while maintaining enteroendocrine L-cells and serum GLP-1 in the normal range. In contrast to an improved glucose tolerance test (GTT) before pregnancy, significantly impaired GTT and remarkably higher serum glucose concentrations in the fed state were observed in α-null dams. Glucagon receptor antagonism treatment, however, did not affect measures of maternal glucose metabolism, indicating a dispensable role of glucagon receptor signaling in maternal glucose homeostasis. However, the GLP-1 receptor agonist improved insulin production and glucose metabolism of α-null dams. Furthermore, GLP-1 receptor antagonist Exendin (9-39) attenuated pregnancy-enhanced insulin secretion and GLP-1 restored glucose-induced insulin secretion of cultured islets from α-null dams. Together, these results demonstrate that α-cells play an essential role in controlling maternal metabolic adaptation to pregnancy by enhancing insulin secretion.
Collapse
Affiliation(s)
- Liping Qiao
- Department of Pediatrics, University of California, San Diego, La Jolla, CA
| | - Sarah Saget
- Department of Pediatrics, University of California, San Diego, La Jolla, CA
| | - Cindy Lu
- Department of Pediatrics, University of California, San Diego, La Jolla, CA
| | - Tianyi Zang
- Department of Pediatrics, University of California, San Diego, La Jolla, CA
| | - Brianna Dzyuba
- Department of Pediatrics, University of California, San Diego, La Jolla, CA
| | | | - Jianhua Shao
- Department of Pediatrics, University of California, San Diego, La Jolla, CA
- Corresponding author: Jianhua Shao,
| |
Collapse
|
37
|
Jia S, Zhao F. Ex vivo development of the entire mouse fetal reproductive tract by using microdissection and membrane-based organ culture techniques. Differentiation 2022; 123:42-49. [PMID: 35030420 PMCID: PMC8821157 DOI: 10.1016/j.diff.2022.01.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 12/13/2021] [Accepted: 01/03/2022] [Indexed: 01/09/2023]
Abstract
Ex vivo explant culture is an appealing alternative to in vivo studies on fetal reproductive organ development. There is extensive literature on ex vivo methods of growing the fetal gonad. However, a method for culturing the whole fetal reproductive tract that has a different shape and size has not been documented. Here, with careful dissection and proper tissue orientation, we successfully cultured the entire bicornuate reproductive tracts from mouse embryos of both sexes on the Transwell insert membrane. The cultured reproductive tract system undergoes sexually dimorphic establishment and region-specific morphogenesis comparable to in vivo development of their counterparts. To test this culture method's applications, we used chemical treatment (dihydrotestosterone and BMS 564929) and genetic cellular ablation mouse model (Gli1-CreER; Rosa-DTA) to investigate the roles of androgen signaling and Gli1+ mesenchyme in Wolffian duct development. Dihydrotestosterone and BMS 564929 promoted the ectopic maintenance of Wolffian ducts in cultured XX tissues. The efficient and specific elimination of Gli1+ mesenchyme was successfully achieved in the cultured tissues, resulting in defective coiling of Wolffian ducts. These results demonstrate the amenability of this organ culture method for chemical and genetic manipulations that are otherwise difficult to study in vivo. Taken together, the establishment of this organ culture method provides a valuable tool complementary to in vivo studies for understanding fetal reproductive tract development in mice.
Collapse
Affiliation(s)
- Shuai Jia
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Fei Zhao
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA,Corresponding author: Fei Zhao, Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA, Tel: 608-890-2610.
| |
Collapse
|
38
|
George KK, Heithoff BP, Shandra O, Robel S. Mild Traumatic Brain Injury/Concussion Initiates an Atypical Astrocyte Response Caused by Blood-Brain Barrier Dysfunction. J Neurotrauma 2022; 39:211-226. [PMID: 34806422 PMCID: PMC8785769 DOI: 10.1089/neu.2021.0204] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Mild traumatic brain injury/concussion (mTBI) accounts for 70-90% of all reported TBI cases and causes long-lasting neurological consequences in 10-40% of patients. Recent clinical studies revealed increased blood-brain barrier (BBB) permeability in mTBI patients, which correlated with secondary damage after mTBI. However, the cascade of cellular events initiated by exposure to blood-borne factors resulting in sustained damage is not fully understood. We previously reported that astrocytes respond atypically to mTBI, rapidly losing many proteins essential to their homeostatic function, while classic scar formation does not occur. Here, we tested the hypothesis that mTBI-induced BBB damage causes atypical astrocytes through exposure to blood-borne factors. Using an mTBI mouse model, two-photon imaging, an endothelial cell-specific genetic ablation approach, and serum-free primary astrocyte cultures, we demonstrated that areas with atypical astrocytes coincide with BBB damage and that exposure of astrocytes to plasma proteins is sufficient to initiate loss of astrocyte homeostatic proteins. Although mTBI resulted in frequent impairment of both physical and metabolic BBB properties and leakage of small-sized blood-borne factors, deposition of the coagulation factor fibrinogen or vessel rupture were rare. Surprisingly, even months after mTBI, BBB repair did not occur in areas with atypical astrocytes. Together, these findings implicate that even relatively small BBB disturbances are sustained long term, and render nearby astrocytes dysfunctional, likely at the cost of neuronal health and function.
Collapse
Affiliation(s)
- Kijana K. George
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, USA.,Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Roanoke, Virginia, USA
| | - Benjamin P. Heithoff
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, USA.,Department of Biological Sciences, Virginia Tech, Blacksburg, Virginia, USA
| | - Oleksii Shandra
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, USA.,Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Stefanie Robel
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, Virginia, USA.,Department of Biological Sciences, Virginia Tech, Blacksburg, Virginia, USA.,Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA.,Address correspondence to: Stefanie Robel, PhD, University of Alabama at Birmingham, Department of Cell, Developmental, and Integrative Biology, McCallum Basic Health Sciences Building, 9th floor, 1720 2nd Avenue South, Birmingham, AL 35294-0006, USA
| |
Collapse
|
39
|
Udagawa T, Atkinson PJ, Milon B, Abitbol JM, Song Y, Sperber M, Huarcaya Najarro E, Scheibinger M, Elkon R, Hertzano R, Cheng AG. Lineage-tracing and translatomic analysis of damage-inducible mitotic cochlear progenitors identifies candidate genes regulating regeneration. PLoS Biol 2021; 19:e3001445. [PMID: 34758021 PMCID: PMC8608324 DOI: 10.1371/journal.pbio.3001445] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 11/22/2021] [Accepted: 10/18/2021] [Indexed: 12/21/2022] Open
Abstract
Cochlear supporting cells (SCs) are glia-like cells critical for hearing function. In the neonatal cochlea, the greater epithelial ridge (GER) is a mitotically quiescent and transient organ, which has been shown to nonmitotically regenerate SCs. Here, we ablated Lgr5+ SCs using Lgr5-DTR mice and found mitotic regeneration of SCs by GER cells in vivo. With lineage tracing, we show that the GER houses progenitor cells that robustly divide and migrate into the organ of Corti to replenish ablated SCs. Regenerated SCs display coordinated calcium transients, markers of the SC subtype inner phalangeal cells, and survive in the mature cochlea. Via RiboTag, RNA-sequencing, and gene clustering algorithms, we reveal 11 distinct gene clusters comprising markers of the quiescent and damaged GER, and damage-responsive genes driving cell migration and mitotic regeneration. Together, our study characterizes GER cells as mitotic progenitors with regenerative potential and unveils their quiescent and damaged translatomes.
Collapse
Affiliation(s)
- Tomokatsu Udagawa
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, California, United States of America
- Department of Otorhinolaryngology, The Jikei University School of Medicine, Tokyo, Japan
| | - Patrick J. Atkinson
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, California, United States of America
| | - Beatrice Milon
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Julia M. Abitbol
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, California, United States of America
| | - Yang Song
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Michal Sperber
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Elvis Huarcaya Najarro
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, California, United States of America
| | - Mirko Scheibinger
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, California, United States of America
| | - Ran Elkon
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ronna Hertzano
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Alan G. Cheng
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, California, United States of America
- * E-mail:
| |
Collapse
|
40
|
Jiang L, Chen T, Sun S, Wang R, Deng J, Lyu L, Wu H, Yang M, Pu X, Du L, Chen Q, Hu Y, Hu X, Zhou Y, Xu Q, Zhang L. Nonbone Marrow CD34 + Cells Are Crucial for Endothelial Repair of Injured Artery. Circ Res 2021; 129:e146-e165. [PMID: 34474592 DOI: 10.1161/circresaha.121.319494] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Liujun Jiang
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China (L. Jiang, T. Chen, S. Sun, R. Wang, J. Deng, L. Lyu, H. Wu, X. Pu, L. Du, Y. Hu, X. Hu, Y. Zhou, Q. Xu)
| | - Ting Chen
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China (L. Jiang, T. Chen, S. Sun, R. Wang, J. Deng, L. Lyu, H. Wu, X. Pu, L. Du, Y. Hu, X. Hu, Y. Zhou, Q. Xu).,Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, Zhejiang Province, China (T. Chen)
| | - Shasha Sun
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China (L. Jiang, T. Chen, S. Sun, R. Wang, J. Deng, L. Lyu, H. Wu, X. Pu, L. Du, Y. Hu, X. Hu, Y. Zhou, Q. Xu).,Department of Cardiology and Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China. (S. Sun, M. Yang, Q. Chen, L. Zhang)
| | - Ruilin Wang
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China (L. Jiang, T. Chen, S. Sun, R. Wang, J. Deng, L. Lyu, H. Wu, X. Pu, L. Du, Y. Hu, X. Hu, Y. Zhou, Q. Xu)
| | - Jiacheng Deng
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China (L. Jiang, T. Chen, S. Sun, R. Wang, J. Deng, L. Lyu, H. Wu, X. Pu, L. Du, Y. Hu, X. Hu, Y. Zhou, Q. Xu)
| | - Lingxia Lyu
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China (L. Jiang, T. Chen, S. Sun, R. Wang, J. Deng, L. Lyu, H. Wu, X. Pu, L. Du, Y. Hu, X. Hu, Y. Zhou, Q. Xu)
| | - Hong Wu
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China (L. Jiang, T. Chen, S. Sun, R. Wang, J. Deng, L. Lyu, H. Wu, X. Pu, L. Du, Y. Hu, X. Hu, Y. Zhou, Q. Xu)
| | - Mei Yang
- Department of Cardiology and Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China. (S. Sun, M. Yang, Q. Chen, L. Zhang)
| | - Xiangyuan Pu
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China (L. Jiang, T. Chen, S. Sun, R. Wang, J. Deng, L. Lyu, H. Wu, X. Pu, L. Du, Y. Hu, X. Hu, Y. Zhou, Q. Xu)
| | - Luping Du
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China (L. Jiang, T. Chen, S. Sun, R. Wang, J. Deng, L. Lyu, H. Wu, X. Pu, L. Du, Y. Hu, X. Hu, Y. Zhou, Q. Xu)
| | - Qishan Chen
- Department of Cardiology and Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China. (S. Sun, M. Yang, Q. Chen, L. Zhang)
| | - Yanhua Hu
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China (L. Jiang, T. Chen, S. Sun, R. Wang, J. Deng, L. Lyu, H. Wu, X. Pu, L. Du, Y. Hu, X. Hu, Y. Zhou, Q. Xu)
| | - Xiaosheng Hu
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China (L. Jiang, T. Chen, S. Sun, R. Wang, J. Deng, L. Lyu, H. Wu, X. Pu, L. Du, Y. Hu, X. Hu, Y. Zhou, Q. Xu)
| | - Yijiang Zhou
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China (L. Jiang, T. Chen, S. Sun, R. Wang, J. Deng, L. Lyu, H. Wu, X. Pu, L. Du, Y. Hu, X. Hu, Y. Zhou, Q. Xu)
| | - Qingbo Xu
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China (L. Jiang, T. Chen, S. Sun, R. Wang, J. Deng, L. Lyu, H. Wu, X. Pu, L. Du, Y. Hu, X. Hu, Y. Zhou, Q. Xu).,Centre for Clinical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom (Q. Xu)
| | - Li Zhang
- Department of Cardiology and Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China. (S. Sun, M. Yang, Q. Chen, L. Zhang)
| |
Collapse
|
41
|
Tseng PY, Hoon MA. Specific β-Defensins Stimulate Pruritus through Activation of Sensory Neurons. J Invest Dermatol 2021; 142:594-602. [PMID: 34480893 DOI: 10.1016/j.jid.2021.07.178] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 07/02/2021] [Accepted: 07/30/2021] [Indexed: 12/12/2022]
Abstract
Pruritus is a common symptom of dermatological disorders and has a major negative impact on QOL. Previously, it was suggested that human β-defensin peptides elicit itch through the activation of mast cells. In this study, we investigated in more detail the mechanisms by which β-defensins induce itch by defining the receptors activated by these peptides in humans and mice, by establishing their action in vivo, and by examining their expression in inflammatory dermal diseases. We found that elevated expression of DEFB103 is highly correlated with skin lesions in psoriasis and atopic dermatitis. We showed that the peptide encoded by this gene and related genes activate Mas-related G protein-coupled receptors with different potencies that are related to their charge density. Furthermore, we establish that although these peptides can activate mast cells, they also activate sensory neurons, with the former cells being dispensable for itch reactions in mice. Together, our studies highlight that specific β-defensins are likely endogenous pruritogens that can directly stimulate sensory neurons.
Collapse
Affiliation(s)
- Pang-Yen Tseng
- Molecular Genetics Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Mark A Hoon
- Molecular Genetics Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA.
| |
Collapse
|
42
|
Zhang X, Robles H, Magee L K, Lorenz R M, Wang Z, Harris A C, Craft S C, Scheller L E. A bone-specific adipogenesis pathway in fat-free mice defines key origins and adaptations of bone marrow adipocytes with age and disease. eLife 2021; 10:66275. [PMID: 34378533 PMCID: PMC8412938 DOI: 10.7554/elife.66275] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 08/02/2021] [Indexed: 02/07/2023] Open
Abstract
Bone marrow adipocytes accumulate with age and in diverse disease states. However, their origins and adaptations in these conditions remain unclear, impairing our understanding of their context-specific endocrine functions and relationship with surrounding tissues. In this study, by analyzing bone and adipose tissues in the lipodystrophic ‘fat-free’ mouse, we define a novel, secondary adipogenesis pathway that relies on the recruitment of adiponectin-negative stromal progenitors. This pathway is unique to the bone marrow and is activated with age and in states of metabolic stress in the fat-free mouse model, resulting in the expansion of bone marrow adipocytes specialized for lipid storage with compromised lipid mobilization and cytokine expression within regions traditionally devoted to hematopoiesis. This finding further distinguishes bone marrow from peripheral adipocytes and contributes to our understanding of bone marrow adipocyte origins, adaptations, and relationships with surrounding tissues with age and disease.
Collapse
Affiliation(s)
- Xiao Zhang
- Division of Bone and Mineral Diseases, Department of Medicine, Washington University, Saint Louis, United States.,Department of Biomedical Engineering, Washington University, Saint Louis, United States
| | - Hero Robles
- Division of Bone and Mineral Diseases, Department of Medicine, Washington University, Saint Louis, United States
| | - Kristann Magee L
- Division of Bone and Mineral Diseases, Department of Medicine, Washington University, Saint Louis, United States
| | - Madelyn Lorenz R
- Division of Bone and Mineral Diseases, Department of Medicine, Washington University, Saint Louis, United States
| | - Zhaohua Wang
- Division of Bone and Mineral Diseases, Department of Medicine, Washington University, Saint Louis, United States.,Department of Orthopaedic Surgery, Washington University, Saint Louis, United States
| | - Charles Harris A
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University, Saint Louis, United States
| | - Clarissa Craft S
- Division of Bone and Mineral Diseases, Department of Medicine, Washington University, Saint Louis, United States
| | - Erica Scheller L
- Division of Bone and Mineral Diseases, Department of Medicine, Washington University, Saint Louis, United States.,Department of Biomedical Engineering, Washington University, Saint Louis, United States
| |
Collapse
|
43
|
Elimination of glutamatergic transmission from Hb9 interneurons does not impact treadmill locomotion. Sci Rep 2021; 11:16008. [PMID: 34362940 PMCID: PMC8346588 DOI: 10.1038/s41598-021-95143-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 07/20/2021] [Indexed: 11/08/2022] Open
Abstract
The spinal cord contains neural circuits that can produce the rhythm and pattern of locomotor activity. It has previously been postulated that a population of glutamatergic neurons, termed Hb9 interneurons, contributes to locomotor rhythmogenesis. These neurons were identified by their expression of the homeobox gene, Hb9, which is also expressed in motor neurons. We developed a mouse line in which Cre recombinase activity is inducible in neurons expressing Hb9. We then used this line to eliminate vesicular glutamate transporter 2 from Hb9 interneurons, and found that there were no deficits in treadmill locomotion. We conclude that glutamatergic neurotransmission by Hb9 interneurons is not required for locomotor behaviour. The role of these neurons in neural circuits remains elusive.
Collapse
|
44
|
Wilke JBH, Hindermann M, Moussavi A, Butt UJ, Dadarwal R, Berghoff SA, Sarcheshmeh AK, Ronnenberg A, Zihsler S, Arinrad S, Hardeland R, Seidel J, Lühder F, Nave KA, Boretius S, Ehrenreich H. Inducing sterile pyramidal neuronal death in mice to model distinct aspects of gray matter encephalitis. Acta Neuropathol Commun 2021; 9:121. [PMID: 34215338 PMCID: PMC8253243 DOI: 10.1186/s40478-021-01214-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 06/07/2021] [Indexed: 02/07/2023] Open
Abstract
Up to one person in a population of 10,000 is diagnosed once in lifetime with an encephalitis, in 50-70% of unknown origin. Recognized causes amount to 20-50% viral infections. Approximately one third of affected subjects develops moderate and severe subsequent damage. Several neurotropic viruses can directly infect pyramidal neurons and induce neuronal death in cortex and hippocampus. The resulting encephalitic syndromes are frequently associated with cognitive deterioration and dementia, but involve numerous parallel and downstream cellular and molecular events that make the interpretation of direct consequences of sudden pyramidal neuronal loss difficult. This, however, would be pivotal for understanding how neuroinflammatory processes initiate the development of neurodegeneration, and thus for targeted prophylactic and therapeutic interventions. Here we utilized adult male NexCreERT2xRosa26-eGFP-DTA (= 'DTA') mice for the induction of a sterile encephalitis by diphtheria toxin-mediated ablation of cortical and hippocampal pyramidal neurons which also recruits immune cells into gray matter. We report multifaceted aftereffects of this defined process, including the expected pathology of classical hippocampal behaviors, evaluated in Morris water maze, but also of (pre)frontal circuit function, assessed by prepulse inhibition. Importantly, we modelled in encephalitis mice novel translationally relevant sequelae, namely altered social interaction/cognition, accompanied by compromised thermoreaction to social stimuli as convenient readout of parallel autonomic nervous system (dys)function. High resolution magnetic resonance imaging disclosed distinct abnormalities in brain dimensions, including cortical and hippocampal layering, as well as of cerebral blood flow and volume. Fluorescent tracer injection, immunohistochemistry and brain flow cytometry revealed persistent blood-brain-barrier perturbance and chronic brain inflammation. Surprisingly, blood flow cytometry showed no abnormalities in circulating major immune cell subsets and plasma high-mobility group box 1 (HMGB1) as proinflammatory marker remained unchanged. The present experimental work, analyzing multidimensional outcomes of direct pyramidal neuronal loss, will open new avenues for urgently needed encephalitis research.
Collapse
Affiliation(s)
- Justus B H Wilke
- Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Hermann-Rein-Str.3, 37075, Göttingen, Germany
| | - Martin Hindermann
- Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Hermann-Rein-Str.3, 37075, Göttingen, Germany
| | - Amir Moussavi
- Functional Imaging Laboratory, German Primate Center, Leibniz Institute for Primate Research, Kellnerweg 4, 37077, Göttingen, Germany
| | - Umer Javed Butt
- Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Hermann-Rein-Str.3, 37075, Göttingen, Germany
| | - Rakshit Dadarwal
- Functional Imaging Laboratory, German Primate Center, Leibniz Institute for Primate Research, Kellnerweg 4, 37077, Göttingen, Germany
- Georg August University, Göttingen, Germany
| | - Stefan A Berghoff
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Aref Kalantari Sarcheshmeh
- Functional Imaging Laboratory, German Primate Center, Leibniz Institute for Primate Research, Kellnerweg 4, 37077, Göttingen, Germany
| | - Anja Ronnenberg
- Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Hermann-Rein-Str.3, 37075, Göttingen, Germany
| | - Svenja Zihsler
- Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Hermann-Rein-Str.3, 37075, Göttingen, Germany
| | - Sahab Arinrad
- Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Hermann-Rein-Str.3, 37075, Göttingen, Germany
| | - Rüdiger Hardeland
- Johann Friedrich Blumenbach Institute of Zoology & Anthropology, University of Göttingen, Göttingen, Germany
| | - Jan Seidel
- Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Hermann-Rein-Str.3, 37075, Göttingen, Germany
| | - Fred Lühder
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Göttingen, Göttingen, Germany
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Susann Boretius
- Functional Imaging Laboratory, German Primate Center, Leibniz Institute for Primate Research, Kellnerweg 4, 37077, Göttingen, Germany.
- Georg August University, Göttingen, Germany.
| | - Hannelore Ehrenreich
- Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Hermann-Rein-Str.3, 37075, Göttingen, Germany.
| |
Collapse
|
45
|
Chen J, Li M, Liu AQ, Zheng CX, Bao LH, Chen K, Xu XL, Guan JT, Bai M, Zhou T, Sui BD, Li DH, Jin Y, Hu CH. Gli1 + Cells Couple with Type H Vessels and Are Required for Type H Vessel Formation. Stem Cell Reports 2021; 15:110-124. [PMID: 32668219 PMCID: PMC7363988 DOI: 10.1016/j.stemcr.2020.06.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 06/05/2020] [Accepted: 06/08/2020] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) reside in the perivascular niche and modulate tissue/organ homeostasis; however, little is known about whether and how their localization and function are linked. Particularly, whether specific MSC subsets couple with and regulate specialized vessel subtypes is unclear. Here, we show that Gli1+ cells, which are a subpopulation of MSCs couple with and regulate a specialized form of vasculature. The specific capillaries, i.e., CD31hiEMCNhi type H vessels, are the preferable vascular subtype which Gli1+ cells are adjacent to in bone. Gli1+ cells are further identified to be phenotypically coupled with type H endothelium during bone growth and defect healing. Importantly, Gli1+ cell ablation inhibits type H vessel formation associated with suppressed bone generation and regeneration. Mechanistically, Gli1+ cells initiate angiogenesis through Gli and HIF-1α signaling. These findings suggest a morphological and functional framework of Gli1+ cells modulating coupled type H vasculature for tissue homeostasis and regenerative repair.
Collapse
Affiliation(s)
- Ji Chen
- State Key Laboratory of Military Stomatology& National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, 145 West Changle Road, Xi'an, Shaanxi 710032, China; Department of Oral Implantology, School of Stomatology, Fourth Military Medical University, Xi'an, Shannxi 710032, China
| | - Meng Li
- State Key Laboratory of Military Stomatology& National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, 145 West Changle Road, Xi'an, Shaanxi 710032, China
| | - An-Qi Liu
- State Key Laboratory of Military Stomatology& National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, 145 West Changle Road, Xi'an, Shaanxi 710032, China
| | - Chen-Xi Zheng
- State Key Laboratory of Military Stomatology& National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, 145 West Changle Road, Xi'an, Shaanxi 710032, China
| | - Li-Hui Bao
- State Key Laboratory of Military Stomatology& National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, 145 West Changle Road, Xi'an, Shaanxi 710032, China; Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, Shaanxi 710032, China
| | - Kai Chen
- State Key Laboratory of Military Stomatology& National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, 145 West Changle Road, Xi'an, Shaanxi 710032, China; Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, Shaanxi 710032, China
| | - Xiao-Lin Xu
- State Key Laboratory of Military Stomatology& National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, 145 West Changle Road, Xi'an, Shaanxi 710032, China; Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, Shaanxi 710032, China
| | - Jiang-Tao Guan
- State Key Laboratory of Military Stomatology& National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, 145 West Changle Road, Xi'an, Shaanxi 710032, China; Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, Shaanxi 710032, China
| | - Meng Bai
- State Key Laboratory of Military Stomatology& National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, 145 West Changle Road, Xi'an, Shaanxi 710032, China; Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, Shaanxi 710032, China
| | - Tao Zhou
- State Key Laboratory of Military Stomatology& National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, 145 West Changle Road, Xi'an, Shaanxi 710032, China; Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, Shaanxi 710032, China
| | - Bing-Dong Sui
- State Key Laboratory of Military Stomatology& National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, 145 West Changle Road, Xi'an, Shaanxi 710032, China
| | - De-Hua Li
- Department of Oral Implantology, School of Stomatology, Fourth Military Medical University, Xi'an, Shannxi 710032, China.
| | - Yan Jin
- State Key Laboratory of Military Stomatology& National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, 145 West Changle Road, Xi'an, Shaanxi 710032, China.
| | - Cheng-Hu Hu
- State Key Laboratory of Military Stomatology& National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, 145 West Changle Road, Xi'an, Shaanxi 710032, China; Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, Shaanxi 710032, China.
| |
Collapse
|
46
|
Park S, Matte-Martone C, Gonzalez DG, Lathrop EA, May DP, Pineda CM, Moore JL, Boucher JD, Marsh E, Schmitter-Sánchez A, Cockburn K, Markova O, Bellaïche Y, Greco V. Skin-resident immune cells actively coordinate their distribution with epidermal cells during homeostasis. Nat Cell Biol 2021; 23:476-484. [PMID: 33958758 DOI: 10.1038/s41556-021-00670-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 03/24/2021] [Indexed: 01/12/2023]
Abstract
Organs consist of multiple cell types that ensure proper architecture and function. How different cell types coexist and interact to maintain their homeostasis in vivo remains elusive. The skin epidermis comprises mostly epithelial cells, but also harbours Langerhans cells (LCs) and dendritic epidermal T cells (DETCs). Whether and how distributions of LCs and DETCs are regulated during homeostasis is unclear. Here, by tracking individual cells in the skin of live adult mice over time, we show that LCs and DETCs actively maintain a non-random spatial distribution despite continuous turnover of neighbouring basal epithelial cells. Moreover, the density of epithelial cells regulates the composition of LCs and DETCs in the epidermis. Finally, LCs require the GTPase Rac1 to maintain their positional stability, density and tiling pattern reminiscent of neuronal self-avoidance. We propose that these cellular mechanisms provide the epidermis with an optimal response to environmental insults.
Collapse
Affiliation(s)
- Sangbum Park
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA.,Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI, USA.,Division of Dermatology, Department of Medicine, College of Human Medicine, Michigan State University, East Lansing, MI, USA.,Department of Pharmacology and Toxicology, College of Human Medicine, Michigan State University, East Lansing, MI, USA
| | | | - David G Gonzalez
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
| | | | - Dennis P May
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
| | | | - Jessica L Moore
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
| | | | - Edward Marsh
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
| | - Axel Schmitter-Sánchez
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI, USA.,Cell and Molecular Biology Program, College of Natural Science, Michigan State University, East Lansing, MI, USA
| | - Katie Cockburn
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
| | - Olga Markova
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Yohanns Bellaïche
- Génétique et Biologie du Développement, Institut Curie, Université PSL, CNRS UMR3215, INSERM U934, Paris, France
| | - Valentina Greco
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA. .,Department of Cell Biology, Yale School of Medicine, New Haven, CT, USA. .,Department of Dermatology, Yale School of Medicine, New Haven, CT, USA. .,Yale Stem Cell Center, Yale School of Medicine, New Haven, CT, USA. .,Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
47
|
Jafree DJ, Long DA, Scambler PJ, Ruhrberg C. Mechanisms and cell lineages in lymphatic vascular development. Angiogenesis 2021; 24:271-288. [PMID: 33825109 PMCID: PMC8205918 DOI: 10.1007/s10456-021-09784-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 03/10/2021] [Indexed: 12/20/2022]
Abstract
Lymphatic vessels have critical roles in both health and disease and their study is a rapidly evolving area of vascular biology. The consensus on how the first lymphatic vessels arise in the developing embryo has recently shifted. Originally, they were thought to solely derive by sprouting from veins. Since then, several studies have uncovered novel cellular mechanisms and a diversity of contributing cell lineages in the formation of organ lymphatic vasculature. Here, we review the key mechanisms and cell lineages contributing to lymphatic development, discuss the advantages and limitations of experimental techniques used for their study and highlight remaining knowledge gaps that require urgent attention. Emerging technologies should accelerate our understanding of how lymphatic vessels develop normally and how they contribute to disease.
Collapse
Affiliation(s)
- Daniyal J Jafree
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK
- Faculty of Medical Sciences, University College London, London, UK
| | - David A Long
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK
| | - Peter J Scambler
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK
| | - Christiana Ruhrberg
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK.
| |
Collapse
|
48
|
Hayata Y, Nakagawa H, Kurosaki S, Kawamura S, Matsushita Y, Hayakawa Y, Suzuki N, Hata M, Tsuboi M, Kinoshita H, Miyabayashi K, Mizutani H, Nakagomi R, Ikenoue T, Hirata Y, Arita J, Hasegawa K, Tateishi K, Koike K. Axin2 + Peribiliary Glands in the Periampullary Region Generate Biliary Epithelial Stem Cells That Give Rise to Ampullary Carcinoma. Gastroenterology 2021; 160:2133-2148.e6. [PMID: 33465373 DOI: 10.1053/j.gastro.2021.01.028] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 01/11/2021] [Accepted: 01/11/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND & AIMS Peribiliary glands (PBGs), clusters of epithelial cells residing in the submucosal compartment of extrahepatic bile ducts, have been suggested as biliary epithelial stem/progenitor cell niche; however, evidence to support this claim is limited because of a lack of PBG-specific markers. We therefore sought to identify PBG-specific markers to investigate the potential role of PBGs as stem/progenitor cell niches, as well as an origin of cancer. METHODS We examined the expression pattern of the Wnt target gene Axin2 in extrahepatic bile ducts. We then applied lineage tracing to investigate whether Axin2-expressing cells from PBGs contribute to biliary regeneration and carcinogenesis using Axin2-CreERT mice. RESULTS Wnt signaling activation, marked by Axin2, was limited to PBGs located in the periampullary region. Lineage tracing showed that Axin2-expressing periampullary PBG cells are capable of self-renewal and supplying new biliary epithelial cells (BECs) to the luminal surface. Additionally, the expression pattern of Axin2 and the mature ductal cell marker CK19 were mutually exclusive in periampullary region, and fate tracing of CK19+ luminal surface BECs showed gradual replacement by CK19- cells, further supporting the continuous replenishment of new BECs from PBGs to the luminal surface. We also found that Wnt signal enhancer R-spondin3 secreted from Myh11-expressing stromal cells, corresponding to human sphincter of Oddi, maintained the periampullary Wnt signal-activating niche. Notably, introduction of PTEN deletion into Axin2+ PBG cells, but not CK19+ luminal surface BECs, induced ampullary carcinoma whose development was suppressed by Wnt inhibitor. CONCLUSION A specific cell population receiving Wnt-activating signal in periampullary PBGs functions as biliary epithelial stem/progenitor cells and also the cellular origin of ampullary carcinoma.
Collapse
Affiliation(s)
- Yuki Hayata
- Department of Gastroenterology, The University of Tokyo, Tokyo, Japan
| | - Hayato Nakagawa
- Department of Gastroenterology, The University of Tokyo, Tokyo, Japan.
| | | | - Satoshi Kawamura
- Department of Gastroenterology, The University of Tokyo, Tokyo, Japan
| | - Yuki Matsushita
- Department of Gastroenterology, The University of Tokyo, Tokyo, Japan
| | - Yoku Hayakawa
- Department of Gastroenterology, The University of Tokyo, Tokyo, Japan
| | - Nobumi Suzuki
- Department of Gastroenterology, The University of Tokyo, Tokyo, Japan
| | - Masahiro Hata
- Department of Gastroenterology, The University of Tokyo, Tokyo, Japan
| | - Mayo Tsuboi
- Department of Gastroenterology, The University of Tokyo, Tokyo, Japan
| | - Hiroto Kinoshita
- Department of Gastroenterology, The University of Tokyo, Tokyo, Japan; Division of Gastroenterology, Institute for Adult Diseases, Asahi Life Foundation, Chuo-ku, Tokyo, Japan
| | - Koji Miyabayashi
- Department of Gastroenterology, The University of Tokyo, Tokyo, Japan
| | - Hiroya Mizutani
- Department of Gastroenterology, The University of Tokyo, Tokyo, Japan
| | - Ryo Nakagomi
- Department of Gastroenterology, The University of Tokyo, Tokyo, Japan
| | - Tsuneo Ikenoue
- Division of Clinical Genome Research, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yoshihiro Hirata
- Division of Advanced Genome Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Junichi Arita
- Hepato-Biliary-Pancreatic Surgery Division, Department of Surgery, The University of Tokyo, Tokyo, Japan
| | - Kiyoshi Hasegawa
- Hepato-Biliary-Pancreatic Surgery Division, Department of Surgery, The University of Tokyo, Tokyo, Japan
| | - Keisuke Tateishi
- Department of Gastroenterology, The University of Tokyo, Tokyo, Japan
| | - Kazuhiko Koike
- Department of Gastroenterology, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
49
|
Luu T, Cheung JF, Baccon J, Waldner H. Priming of myelin-specific T cells in the absence of dendritic cells results in accelerated development of Experimental Autoimmune Encephalomyelitis. PLoS One 2021; 16:e0250340. [PMID: 33891644 PMCID: PMC8064509 DOI: 10.1371/journal.pone.0250340] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 04/05/2021] [Indexed: 12/12/2022] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) is an established animal model of multiple sclerosis (MS). Inflammatory CD4+ T cell responses directed against CNS antigens, including myelin proteolipid protein (PLP), are key mediators of EAE. Dendritic cells (DCs) are critical for the induction of T cell responses against infectious agents. However, the importance of DCs in priming self-reactive CD4+ T cells in autoimmune disease such as MS has been unclear. To determine the requirement of DCs in PLP-specific CD4+ T cell responses and EAE, we genetically deleted CD11c+ DCs in PLP T cell receptor (TCR) transgenic SJL mice constitutively. DC deficiency did not impair the development, selection or the pathogenic function of PLP-specific CD4+ T cells in these mice, and resulted in accelerated spontaneous EAE compared to DC sufficient controls. In addition, using a genetic approach to ablate DCs conditionally in SJL mice, we show that CD11c+ DCs were dispensable for presenting exogenous or endogenous myelin antigen to PLP-specific T cells and for promoting pro-inflammatory T cell responses and severe EAE. Our findings demonstrate that constitutive or conditional ablation of CD11c+ DCs diminished self-tolerance to PLP autoantigen. They further show that in the absence of DCs, non-DCs can efficiently present CNS myelin antigens such as PLP to self-reactive T cells, resulting in accelerated onset of spontaneous or induced EAE.
Collapse
Affiliation(s)
- Thaiphi Luu
- Department of Microbiology & Immunology, Penn State College of Medicine, Hershey, Pennsylvania, United States of America
| | - Julie F. Cheung
- Department of Microbiology & Immunology, Penn State College of Medicine, Hershey, Pennsylvania, United States of America
| | - Jennifer Baccon
- Department of Pathology, Penn State College of Medicine, Hershey, Pennsylvania, United States of America
- Department of Neurosurgery, Penn State College of Medicine, Hershey, Pennsylvania, United States of America
| | - Hanspeter Waldner
- Department of Microbiology & Immunology, Penn State College of Medicine, Hershey, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
50
|
Singh R, Ha SE, Wei L, Jin B, Zogg H, Poudrier SM, Jorgensen BG, Park C, Ronkon CF, Bartlett A, Cho S, Morales A, Chung YH, Lee MY, Park JK, Gottfried-Blackmore A, Nguyen L, Sanders KM, Ro S. miR-10b-5p Rescues Diabetes and Gastrointestinal Dysmotility. Gastroenterology 2021; 160:1662-1678.e18. [PMID: 33421511 PMCID: PMC8532043 DOI: 10.1053/j.gastro.2020.12.062] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/18/2020] [Accepted: 12/26/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS Interstitial cells of Cajal (ICCs) and pancreatic β cells require receptor tyrosine kinase (KIT) to develop and function properly. Degeneration of ICCs is linked to diabetic gastroparesis. The mechanisms linking diabetes and gastroparesis are unclear, but may involve microRNA (miRNA)-mediated post-transcriptional gene silencing in KIT+ cells. METHODS We performed miRNA-sequencing analysis from isolated ICCs in diabetic mice and plasma from patients with idiopathic and diabetic gastroparesis. miR-10b-5p target genes were identified and validated in mouse and human cell lines. For loss-of-function studies, we used KIT+ cell-restricted mir-10b knockout mice and KIT+ cell depletion mice. For gain-of-function studies, a synthetic miR-10b-5p mimic was injected in multiple diabetic mouse models. We compared the efficacy of miR-10b-5p mimic treatment vs antidiabetic and prokinetic medicines. RESULTS miR-10b-5p is highly expressed in ICCs from healthy mice, but drastically depleted in ICCs from diabetic mice. A conditional knockout of mir-10b in KIT+ cells or depletion of KIT+ cells in mice leads to degeneration of β cells and ICCs, resulting in diabetes and gastroparesis. miR-10b-5p targets the transcription factor Krüppel-like factor 11 (KLF11), which negatively regulates KIT expression. The miR-10b-5p mimic or Klf11 small interfering RNAs injected into mir-10b knockout mice, diet-induced diabetic mice, and TALLYHO polygenic diabetic mice rescue the diabetes and gastroparesis phenotype for an extended period of time. Furthermore, the miR-10b-5p mimic is more effective in improving glucose homoeostasis and gastrointestinal motility compared with common antidiabetic and prokinetic medications. CONCLUSIONS miR-10b-5p is a key regulator in diabetes and gastrointestinal dysmotility via the KLF11-KIT pathway. Restoration of miR-10b-5p may provide therapeutic benefits for these disorders.
Collapse
Affiliation(s)
- Rajan Singh
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Se Eun Ha
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Lai Wei
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Byungchang Jin
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Hannah Zogg
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Sandra M. Poudrier
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Brian G. Jorgensen
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Chanjae Park
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Charles F Ronkon
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Allison Bartlett
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Sung Cho
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Addison Morales
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Yu Heon Chung
- Division of Biological Sciences, Wonkwang University, Iksan, Chonbuk, Korea
| | - Moon Young Lee
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, 89557, USA,Department of Physiology, Wonkwang Digestive Disease Research Institute and Institute of Wonkwang Medical Science, School of Medicine, Wonkwang University, Iksan, Chonbuk, Korea
| | - Jong Kun Park
- Division of Biological Sciences, Wonkwang University, Iksan, Chonbuk, Korea
| | - Andrés Gottfried-Blackmore
- Division of Gastroenterology & Hepatology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Linda Nguyen
- Division of Gastroenterology & Hepatology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Kenton M. Sanders
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, 89557, USA
| | - Seungil Ro
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, Nevada.
| |
Collapse
|