1
|
Gao W, Liu D, Li H, He Z, Liu Y, Li X, Ma H, Shi W. NIR-II Cyazulene Fluorophores with Adjustable Cycloalkenes and Spectroscopic Properties for Imaging Applications. Adv Healthc Mater 2025; 14:e2404996. [PMID: 39955744 DOI: 10.1002/adhm.202404996] [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: 01/08/2025] [Indexed: 02/17/2025]
Abstract
NIR-II fluorescence imaging has shown broad application prospects in life science because of its deeper tissue penetration and higher spatiotemporal resolution. Developing NIR-II fluorophores with special spectroscopic properties and analytical functions has become a cutting-edge but challenging topic in this field. Herein, a series of azulene-based NIR-II fluorophores (called cyazulenes) with adjustable cycloalkenes and spectroscopic properties for imaging applications is reported. Cyazulenes (CA965, CA985, and CA1025) are constructed via the convenient coupling of cycloalkenes with azulene derivatives. They exhibit different H-aggregation and J-aggregation. Particularly noteworthy is that CA985 can form significant J-aggregates with strong and sharp NIR-II emission in biological media, and can be used for in vivo imaging of blood vessels and fine structures of collecting lymphatics in mice. Moreover, CA985 displays passive bone-targeted capacity, thereby enabling bone imaging. This study demonstrates the chemical impact of varying cycloalkenes on the aggregate formation and spectral properties of cyazulenes, which will advance azulene-based NIR-II fluorophores.
Collapse
Affiliation(s)
- Wenjie Gao
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Diankai Liu
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - He Li
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Zixu He
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Ya Liu
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Xiaohua Li
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Huimin Ma
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Wen Shi
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- University of the Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
2
|
Arroyo-Ataz G, Yagüe AC, Breda JC, Mazzilli SA, Jones D. Transcriptional, developmental, and functional parallels of lymphatic and venous smooth muscle. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.18.604042. [PMID: 39091770 PMCID: PMC11291064 DOI: 10.1101/2024.07.18.604042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Lymphatic muscle cells (LMCs) are indispensable for lymphatic vessel contraction and their aberrant recruitment or absence is associated with both primary and secondary lymphedema. Despite their critical role in lymphatic vessel function, the transcriptomic and developmental basis that confer the unique contractile properties to LMCs are largely undefined. In this study, we employed single-cell RNA sequencing (scRNAseq), lineage tracing and in vivo imaging to investigate the basis for the hybrid cardiomyocyte and blood vascular smooth muscle cell (SMC) characteristics that have been described for LMCs. Using scRNAseq, the transcriptomes of LMC and venous SMCs from the murine hindlimb exhibited more similarities than differences, although both were markedly distinct from that of arteriole SMCs in the same tissue. Functionally, both lymphatic vessels and blood vessels in the murine hindlimb displayed pulsatile contractility. However, despite expressing genes that overlap with the venous SMC transcriptome, through lineage tracing we show that LMCs do not originate from Myh11+ SMC progenitors. Previous studies have shown that LMCs express cardiac-related genes, whereas in our study we found that arteriole SMCs, but not LMCs, expressed cardiac-related genes. Through lineage tracing, we demonstrate that a subpopulation of LMCs and SMCs originate from WT1+ mesodermal progenitors, which are known to give rise to SMCs. LMCs, however, do not derive from Nkx2.5+ cardiomyocyte progenitors. Overall, our findings suggest that venous SMCs and LMCs and may derive from a related mesodermal progenitor and adopt a similar gene expression program that enable their contractile properties.
Collapse
Affiliation(s)
- Guillermo Arroyo-Ataz
- Department of Pathology & Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, 670 Albany Street, Boston, Massachusetts 02118, USA
| | - Alejandra Carrasco Yagüe
- Department of Pathology & Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, 670 Albany Street, Boston, Massachusetts 02118, USA
| | - Julia C. Breda
- Department of Medicine, Division of Computational Biomedicine, Boston University Chobanian & Avedisian School of Medicine, 75 E. Newton Street, Boston, Massachusetts 02118, USA
| | - Sarah A. Mazzilli
- Department of Medicine, Division of Computational Biomedicine, Boston University Chobanian & Avedisian School of Medicine, 75 E. Newton Street, Boston, Massachusetts 02118, USA
| | - Dennis Jones
- Department of Pathology & Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, 670 Albany Street, Boston, Massachusetts 02118, USA
| |
Collapse
|
3
|
Davis MJ, Castorena-Gonzalez JA, Li M, Zawieja SD, Simon AM, Geng X, Srinivasan RS. Connexin-45 is expressed in mouse lymphatic endothelium and required for lymphatic valve function. JCI Insight 2024; 9:e169931. [PMID: 39074069 PMCID: PMC11343601 DOI: 10.1172/jci.insight.169931] [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: 03/13/2023] [Accepted: 07/11/2024] [Indexed: 07/31/2024] Open
Abstract
The expression and functional relevance of the gap junction molecule connexin-45 (Cx45; GJC1) in lymphatic endothelium were not previously known. We found that Cx45 was expressed widely in the endothelium of murine lymphatics, in both valve and nonvalve regions. Cell-specific deletion of Cx45, driven by a constitutive Cre line (Lyve1-Cre) or an inducible Cre line (Prox1-CreERT2), compromised the function of lymphatic valves, as assessed by physiological tests (back leak and closure) of isolated, single-valve vessel segments. The defects were comparable to those previously reported for loss of Cx43, and as with Cx43, deletion of Cx45 resulted in shortening or increased asymmetry of lymphatic valve leaflets, providing an explanation for the compromised valve function. In contrast with Cx43, lymphatic endothelial cell-specific (LEC-specific) deletion of Cx45 did not alter the number of valves in mesenteric or dermal lymphatic networks or the expression patterns of the canonical valve-associated proteins PROX1, ITGA9, or CLAUDIN5. Constitutive deletion of Cx45 from LECs resulted in increased backflow of injected tracer in popliteal networks in vivo and compromised the integrity of the LEC permeability barrier in a subset of collecting vessels. These findings provide evidence for an unexpected role of Cx45 in the development and maintenance of lymphatic valves.
Collapse
Affiliation(s)
- Michael J. Davis
- Department of Medical Pharmacology & Physiology, University of Missouri, Columbia, Missouri, USA
| | | | - Min Li
- Department of Medical Pharmacology & Physiology, University of Missouri, Columbia, Missouri, USA
| | - Scott D. Zawieja
- Department of Medical Pharmacology & Physiology, University of Missouri, Columbia, Missouri, USA
| | - Alex M. Simon
- Department of Physiology, University of Arizona School of Medicine, Tucson, Arizona, USA
| | - Xin Geng
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - R. Sathish Srinivasan
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| |
Collapse
|
4
|
Mathur T, Tronolone JJ, Jain A. AngioMT: A MATLAB based 2D image-to-physics tool to predict oxygen transport in vascularized microphysiological systems. PLoS One 2024; 19:e0299160. [PMID: 38748761 PMCID: PMC11095698 DOI: 10.1371/journal.pone.0299160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 02/06/2024] [Indexed: 05/19/2024] Open
Abstract
Microphysiological models (MPS) are increasingly getting recognized as in vitro preclinical systems of pathophysiology and drug discovery. However, there is also a growing need to adapt and advance MPS to include the physiological contributions of the capillary vascular dynamics, because they undergo angiogenesis or vasculogenesis to deliver soluble oxygen and nutrients to its organs. Currently, the process of formation of microvessels in MPS is measured arbitrarily, and vascularized MPS do not include oxygen measurements in their analysis. Sensing and measuring tissue oxygen delivery is extremely difficult because it requires access to opaque and deep tissue, and/or requires extensive integration of biosensors that makes such systems impractical to use in the real world. Here, a finite element method-based oxygen transport program, called AngioMT, is built in MATLAB. AngioMT processes the routinely acquired 2D confocal images of microvascular networks in vitro and solves physical equations of diffusion-reaction dominated oxygen transport phenomena. This user-friendly image-to-physics transition in AngioMT is an enabling tool of MPS analysis because unlike the averaged morphological measures of vessels, it provides information of the spatial transport of oxygen both within the microvessels and the surrounding tissue regions. Further, it solves the more complex higher order reaction mechanisms which also improve the physiological relevance of this tool when compared directly against in vivo measurements. Finally, the program is applied in a multicellular vascularized MPS by including the ability to define additional organ/tissue subtypes in complex co-cultured systems. Therefore, AngioMT serves as an analytical tool to enhance the predictive power and performance of MPS that incorporate microcirculation.
Collapse
Affiliation(s)
- Tanmay Mathur
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, United States of America
| | - James J. Tronolone
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, United States of America
| | - Abhishek Jain
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, United States of America
- Department of Medical Physiology, College of Medicine, Texas A&M Health Science Center, Bryan, Texas, United States of America
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, United States of America
| |
Collapse
|
5
|
K. C. R, Patel NR, Shenoy A, Scallan JP, Chiang MY, Galazo MJ, Meadows SM. Zmiz1 is a novel regulator of lymphatic endothelial cell gene expression and function. PLoS One 2024; 19:e0302926. [PMID: 38718095 PMCID: PMC11078365 DOI: 10.1371/journal.pone.0302926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 04/15/2024] [Indexed: 05/12/2024] Open
Abstract
Zinc Finger MIZ-Type Containing 1 (Zmiz1), also known as ZIMP10 or RAI17, is a transcription cofactor and member of the Protein Inhibitor of Activated STAT (PIAS) family of proteins. Zmiz1 is critical for a variety of biological processes including vascular development. However, its role in the lymphatic vasculature is unknown. In this study, we utilized human dermal lymphatic endothelial cells (HDLECs) and an inducible, lymphatic endothelial cell (LEC)-specific Zmiz1 knockout mouse model to investigate the role of Zmiz1 in LECs. Transcriptional profiling of ZMIZ1-deficient HDLECs revealed downregulation of genes crucial for lymphatic vessel development. Additionally, our findings demonstrated that loss of Zmiz1 results in reduced expression of proliferation and migration genes in HDLECs and reduced proliferation and migration in vitro. We also presented evidence that Zmiz1 regulates Prox1 expression in vitro and in vivo by modulating chromatin accessibility at Prox1 regulatory regions. Furthermore, we observed that loss of Zmiz1 in mesenteric lymphatic vessels significantly reduced valve density. Collectively, our results highlight a novel role of Zmiz1 in LECs and as a transcriptional regulator of Prox1, shedding light on a previously unknown regulatory factor in lymphatic vascular biology.
Collapse
Affiliation(s)
- Rajan K. C.
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, United States of America
| | - Nehal R. Patel
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, United States of America
| | - Anoushka Shenoy
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, United States of America
| | - Joshua P. Scallan
- Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States of America
| | - Mark Y. Chiang
- Department of Internal Medicine, Division of Hematology-Oncology, Medical School, University of Michigan, Ann Arbor, MI, United States of America
| | - Maria J. Galazo
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, United States of America
- Tulane Brain Institute, Tulane University, New Orleans, LA, United States of America
| | - Stryder M. Meadows
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, United States of America
- Tulane Brain Institute, Tulane University, New Orleans, LA, United States of America
| |
Collapse
|
6
|
Carlantoni C, Liekfeld LMH, Hemkemeyer SA, Schreier D, Saygi C, Kurelic R, Cardarelli S, Kalucka J, Schulte C, Beerens M, Mailer RK, Schäffer TE, Naro F, Pellegrini M, Nikolaev VO, Renné T, Frye M. The phosphodiesterase 2A controls lymphatic junctional maturation via cGMP-dependent notch signaling. Dev Cell 2024; 59:308-325.e11. [PMID: 38159569 DOI: 10.1016/j.devcel.2023.12.002] [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: 01/05/2023] [Revised: 11/01/2023] [Accepted: 12/07/2023] [Indexed: 01/03/2024]
Abstract
The molecular mechanisms by which lymphatic vessels induce cell contact inhibition are not understood. Here, we identify the cGMP-dependent phosphodiesterase 2A (PDE2A) as a selective regulator of lymphatic but not of blood endothelial contact inhibition. Conditional deletion of Pde2a in mouse embryos reveals severe lymphatic dysplasia, whereas blood vessel architecture remains unaltered. In the absence of PDE2A, human lymphatic endothelial cells fail to induce mature junctions and cell cycle arrest, whereas cGMP levels, but not cAMP levels, are increased. Loss of PDE2A-mediated cGMP hydrolysis leads to the activation of p38 signaling and downregulation of NOTCH signaling. However, DLL4-induced NOTCH activation restores junctional maturation and contact inhibition in PDE2A-deficient human lymphatic endothelial cells. In postnatal mouse mesenteries, PDE2A is specifically enriched in collecting lymphatic valves, and loss of Pde2a results in the formation of abnormal valves. Our data demonstrate that PDE2A selectively finetunes a crosstalk of cGMP, p38, and NOTCH signaling during lymphatic vessel maturation.
Collapse
Affiliation(s)
- Claudia Carlantoni
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany; German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg/Luebeck/Kiel, Hamburg, Germany
| | - Leon M H Liekfeld
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Sandra A Hemkemeyer
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany; German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg/Luebeck/Kiel, Hamburg, Germany
| | - Danny Schreier
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Ceren Saygi
- Bioinformatics Core, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Roberta Kurelic
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Silvia Cardarelli
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, 00161 Rome, Italy
| | - Joanna Kalucka
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Christian Schulte
- German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg/Luebeck/Kiel, Hamburg, Germany; Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Manu Beerens
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany; German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg/Luebeck/Kiel, Hamburg, Germany
| | - Reiner K Mailer
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Tilman E Schäffer
- Institute of Applied Physics, University of Tuebingen, 72076 Tuebingen, Germany
| | - Fabio Naro
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, 00161 Rome, Italy
| | - Manuela Pellegrini
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, 00161 Rome, Italy; Institute of Biochemistry and Cell Biology, IBBC-CNR, Campus A. Buzzati Traverso, Monterotondo Scalo, Rome 00015, Italy
| | - Viacheslav O Nikolaev
- German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg/Luebeck/Kiel, Hamburg, Germany; Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Thomas Renné
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany; Center for Thrombosis and Hemostasis (CTH), Johannes Gutenberg University Medical Center, Mainz, Germany; Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Maike Frye
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany; German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg/Luebeck/Kiel, Hamburg, Germany.
| |
Collapse
|
7
|
Dave M, Dev A, Somoza RA, Zhao N, Viswanath S, Mina PR, Chirra P, Obmann VC, Mahabeleshwar GH, Menghini P, Durbin-Johnson B, Nolta J, Soto C, Osme A, Khuat LT, Murphy WJ, Caplan AI, Cominelli F. MSCs mediate long-term efficacy in a Crohn's disease model by sustained anti-inflammatory macrophage programming via efferocytosis. NPJ Regen Med 2024; 9:6. [PMID: 38245543 PMCID: PMC10799947 DOI: 10.1038/s41536-024-00347-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 01/04/2024] [Indexed: 01/22/2024] Open
Abstract
Mesenchymal stem cells (MSCs) are novel therapeutics for the treatment of Crohn's disease. However, their mechanism of action is unclear, especially in disease-relevant chronic models of inflammation. Thus, we used SAMP-1/YitFc (SAMP), a chronic and spontaneous murine model of small intestinal inflammation, to study the therapeutic effects and mechanism of action of human bone marrow-derived MSCs (hMSC). hMSC dose-dependently inhibited naïve T lymphocyte proliferation via prostaglandin E2 (PGE2) secretion and reprogrammed macrophages to an anti-inflammatory phenotype. We found that the hMSCs promoted mucosal healing and immunologic response early after administration in SAMP when live hMSCs are present (until day 9) and resulted in a complete response characterized by mucosal, histological, immunologic, and radiological healing by day 28 when no live hMSCs are present. hMSCs mediate their effect via modulation of T cells and macrophages in the mesentery and mesenteric lymph nodes (mLN). Sc-RNAseq confirmed the anti-inflammatory phenotype of macrophages and identified macrophage efferocytosis of apoptotic hMSCs as a mechanism that explains their long-term efficacy. Taken together, our findings show that hMSCs result in healing and tissue regeneration in a chronic model of small intestinal inflammation and despite being short-lived, exert long-term effects via sustained anti-inflammatory programming of macrophages via efferocytosis.
Collapse
Affiliation(s)
- Maneesh Dave
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, UC Davis Medical Center, University of California Davis School of Medicine, Sacramento, CA, USA.
- Institute for Regenerative Cures, University of California Davis School of Medicine, Sacramento, CA, USA.
| | - Atul Dev
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, UC Davis Medical Center, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Rodrigo A Somoza
- Skeletal Research Center, Department of Biology, Case Western Reserve University, Cleveland, OH, USA
| | - Nan Zhao
- Division of Gastroenterology and Liver Disease, University Hospitals, Case Western Reserve University, Cleveland, OH, USA
| | - Satish Viswanath
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Pooja Rani Mina
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, UC Davis Medical Center, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Prathyush Chirra
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Verena Carola Obmann
- Department of Diagnostic, Interventional and Pediatric Radiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Ganapati H Mahabeleshwar
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Paola Menghini
- Division of Gastroenterology and Liver Disease, University Hospitals, Case Western Reserve University, Cleveland, OH, USA
| | - Blythe Durbin-Johnson
- Division of Biostatistics, Department of Public Health Sciences, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Jan Nolta
- Institute for Regenerative Cures, University of California Davis School of Medicine, Sacramento, CA, USA
- Division of Malignant Hematology/Cell and Marrow Transplantation, Department of Internal Medicine, University of California Davis School of Medicine, Sacramento, USA
| | - Christopher Soto
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, UC Davis Medical Center, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Abdullah Osme
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Lam T Khuat
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA, USA
| | - William J Murphy
- Division of Malignant Hematology/Cell and Marrow Transplantation, Department of Internal Medicine, University of California Davis School of Medicine, Sacramento, USA
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Arnold I Caplan
- Skeletal Research Center, Department of Biology, Case Western Reserve University, Cleveland, OH, USA
| | - Fabio Cominelli
- Division of Gastroenterology and Liver Disease, University Hospitals, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
8
|
Rajan KC, Patel NR, Shenoy A, Scallan JP, Chiang MY, Galazo MJ, Meadows SM. Zmiz1 is a novel regulator of lymphatic endothelial cell gene expression and function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.22.550165. [PMID: 37503058 PMCID: PMC10370198 DOI: 10.1101/2023.07.22.550165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Zinc Finger MIZ-Type Containing 1 (Zmiz1), also known as ZIMP10 or RAI17, is a transcription cofactor and member of the Protein Inhibitor of Activated STAT (PIAS) family of proteins. Zmiz1 is critical for a variety of biological processes including vascular development. However, its role in the lymphatic vasculature is unknown. In this study, we utilized human dermal lymphatic endothelial cells (HDLECs) and an inducible, lymphatic endothelial cell (LEC)-specific Zmiz1 knockout mouse model to investigate the role of Zmiz1 in LECs. Transcriptional profiling of Zmiz1-deficient HDLECs revealed downregulation of genes crucial for lymphatic vessel development. Additionally, our findings demonstrated that loss of Zmiz1 results in reduced expression of proliferation and migration genes in HDLECs and reduced proliferation and migration in vitro. We also presented evidence that Zmiz1 regulates Prox1 expression in vitro and in vivo by modulating chromatin accessibility at Prox1 regulatory regions. Furthermore, we observed that loss of Zmiz1 in mesenteric lymphatic vessels significantly reduced valve density. Collectively, our results highlight a novel role of Zmiz1 in LECs and as a transcriptional regulator of Prox1, shedding light on a previously unknown regulatory factor in lymphatic vascular biology.
Collapse
Affiliation(s)
- K C Rajan
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA
| | - Nehal R Patel
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA
| | - Anoushka Shenoy
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA
| | - Joshua P Scallan
- Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Mark Y Chiang
- Division of Hematology-Oncology, Department of Internal Medicine, Medical School, University of Michigan, Ann Arbor, MI
| | - Maria J Galazo
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA
- Tulane Brain Institute, Tulane University, New Orleans, LA
| | - Stryder M Meadows
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA
- Tulane Brain Institute, Tulane University, New Orleans, LA
| |
Collapse
|
9
|
Saygili Demir C, Sabine A, Gong M, Dormond O, Petrova TV. Mechanosensitive mTORC1 signaling maintains lymphatic valves. J Cell Biol 2023; 222:e202207049. [PMID: 37036444 PMCID: PMC10097975 DOI: 10.1083/jcb.202207049] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 01/26/2023] [Accepted: 03/23/2023] [Indexed: 04/11/2023] Open
Abstract
Homeostatic maintenance and repair of lymphatic vessels are essential for health. We investigated the dynamics and the molecular mechanisms of lymphatic endothelial cell (LEC) renewal in adult mesenteric quiescent lymphatic vasculature using label-retention, lineage tracing, and cell ablation strategies. Unlike during development, adult LEC turnover and proliferation was confined to the valve regions of collecting vessels, with valve cells displaying the shortest lifespan. Proliferating valve sinus LECs were the main source for maintenance and repair of lymphatic valves. We identified mechanistic target of rapamycin complex 1 (mTORC1) as a mechanoresponsive pathway activated by fluid shear stress in LECs. Depending on the shear stress level, mTORC1 activity drives division of valve cells or dictates their mechanic resilience through increased protein synthesis. Overactivation of lymphatic mTORC1 in vivo promoted supernumerary valve formation. Our work provides insights into the molecular mechanisms of maintenance of healthy lymphatic vascular system.
Collapse
Affiliation(s)
- Cansaran Saygili Demir
- Department of Oncology, Lausanne University Hospital-University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
| | - Amélie Sabine
- Department of Oncology, Lausanne University Hospital-University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
| | - Muyun Gong
- Department of Oncology, Lausanne University Hospital-University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
| | - Olivier Dormond
- Department of Visceral Surgery, Lausanne University Hospital, Lausanne, Switzerland
| | - Tatiana V. Petrova
- Department of Oncology, Lausanne University Hospital-University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
- Swiss Institute for Experimental Cancer Research, École polytechnique fédérale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
10
|
Dave M, Dev A, Somoza RA, Zhao N, Viswanath S, Mina PR, Chirra P, Obmann VC, Mahabeleshwar GH, Menghini P, Johnson BD, Nolta J, Soto C, Osme A, Khuat LT, Murphy W, Caplan AI, Cominelli F. Mesenchymal stem cells ameliorate inflammation in an experimental model of Crohn's disease via the mesentery. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.22.541829. [PMID: 37292753 PMCID: PMC10245893 DOI: 10.1101/2023.05.22.541829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Objective Mesenchymal stem cells (MSCs) are novel therapeutics for treatment of Crohn's disease. However, their mechanism of action is unclear, especially in disease-relevant chronic models of inflammation. Thus, we used SAMP-1/YitFc, a chronic and spontaneous murine model of small intestinal inflammation, to study the therapeutic effect and mechanism of human bone marrow-derived MSCs (hMSC). Design hMSC immunosuppressive potential was evaluated through in vitro mixed lymphocyte reaction, ELISA, macrophage co-culture, and RT-qPCR. Therapeutic efficacy and mechanism in SAMP were studied by stereomicroscopy, histopathology, MRI radiomics, flow cytometry, RT-qPCR, small animal imaging, and single-cell RNA sequencing (Sc-RNAseq). Results hMSC dose-dependently inhibited naïve T lymphocyte proliferation in MLR via PGE 2 secretion and reprogrammed macrophages to an anti-inflammatory phenotype. hMSC promoted mucosal healing and immunologic response early after administration in SAMP model of chronic small intestinal inflammation when live hMSCs are present (until day 9) and resulted in complete response characterized by mucosal, histological, immunologic, and radiological healing by day 28 when no live hMSCs are present. hMSC mediate their effect via modulation of T cells and macrophages in the mesentery and mesenteric lymph nodes (mLN). Sc-RNAseq confirmed the anti-inflammatory phenotype of macrophages and identified macrophage efferocytosis of apoptotic hMSCs as a mechanism of action that explains their long-term efficacy. Conclusion hMSCs result in healing and tissue regeneration in a chronic model of small intestinal inflammation. Despite being short-lived, exert long-term effects via macrophage reprogramming to an anti-inflammatory phenotype. Data Transparency Statement Single-cell RNA transcriptome datasets are deposited in an online open access repository 'Figshare' (DOI: https://doi.org/10.6084/m9.figshare.21453936.v1 ).
Collapse
|
11
|
Banerjee P, Roy S, Chakraborty S. Recent advancement of imaging strategies of the lymphatic system: Answer to the decades old questions. Microcirculation 2022; 29:e12780. [PMID: 35972391 DOI: 10.1111/micc.12780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 07/22/2022] [Accepted: 08/10/2022] [Indexed: 12/30/2022]
Abstract
The role of the lymphatic system in maintaining tissue homeostasis and a number of different pathophysiological conditions has been well established. The complex and delicate structure of the lymphatics along with the limitations of conventional imaging techniques make lymphatic imaging particularly difficult. Thus, in-depth high-resolution imaging of lymphatic system is key to understanding the progression of lymphatic diseases and cancer metastases and would greatly benefit clinical decisions. In recent years, the advancement of imaging technologies and development of new tracers suitable for clinical applications has enabled imaging of the lymphatic system in both clinical and pre-clinical settings. In this current review, we have highlighted the advantages and disadvantages of different modern techniques such as near infra-red spectroscopy (NIRS), positron emission tomography (PET), computed tomography (CT), magnetic resonance imaging (MRI) and fluorescence optical imaging, that has significantly impacted research in this field and has led to in-depth insights into progression of pathological states. This review also highlights the use of current imaging technologies, and tracers specific for immune cell markers to identify and track the immune cells in the lymphatic system that would help understand disease progression and remission in immune therapy regimen.
Collapse
Affiliation(s)
- Priyanka Banerjee
- Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Sukanya Roy
- Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Sanjukta Chakraborty
- Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas, USA
| |
Collapse
|
12
|
Molina SA, Maier-Begandt D, Isakson BE, Koval M. Electrophysiological Measurements of Isolated Blood Vessels. Bio Protoc 2022; 12:e4359. [PMID: 35434187 PMCID: PMC8983162 DOI: 10.21769/bioprotoc.4359] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 11/12/2021] [Accepted: 02/07/2022] [Indexed: 10/07/2023] Open
Abstract
The lumen of blood vessels is covered by endothelial cells, which regulate their permeability to ions and solutes. Endothelial permeability depends on the vascular bed and cell phenotype, and is influenced by different disease states. Most characterization of endothelial permeability has been carried out using isolated cells in culture. While analysis of cultured cells is a valuable approach, it does not account for factors of the native cell environment. Building on Ussing chamber studies of intact tissue specimens, here we describe a method to measure the electrophysiological properties of intact arteriole and venule endothelia, including transendothelial electrical resistance (TEER) and ion permselectivity. As an example, vessels isolated from the mesentery were treated ex vivo, then mounted in a custom-made tissue cassette that enable their analysis by classical approaches with an Ussing chamber. This method enables a detailed analysis of electrophysiological vessel responses to stresses such as proinflammatory cytokines, in the context of an intact vessel. Graphic abstract.
Collapse
Affiliation(s)
- Samuel A Molina
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Daniela Maier-Begandt
- Robert M. Berne Cardiovascular Research Center, University of Virginia, School of Medicine, Charlottesville, VA 22908, USA
- Walter Brendel Center of Experimental Medicine, University Hospital, and Institute of Cardiovascular Physiology and Pathophysiology, Biomedical Center, LMU Munich, 82152 Planegg-Martinsried, Germany
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia, School of Medicine, Charlottesville, VA 22908, USA
- Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Michael Koval
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
13
|
Castorena-Gonzalez JA. Lymphatic Valve Dysfunction in Western Diet-Fed Mice: New Insights Into Obesity-Induced Lymphedema. Front Pharmacol 2022; 13:823266. [PMID: 35308249 PMCID: PMC8931217 DOI: 10.3389/fphar.2022.823266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 02/14/2022] [Indexed: 11/13/2022] Open
Abstract
A two-way connection between obesity and lymphatic dysfunction has now been established. Clinical studies have demonstrated that obesity significantly increases the risk for developing secondary lymphedema. Using animal-models, obesity and metabolic syndrome have been linked to different aspects of lymphatic structural abnormalities and lymphatic dysfunction, including impaired contractility, impaired flow-mediated responses, impaired fluid transport, as well as increased permeability, and abnormal dendritic cell migration among others. Dysfunction of lymphatic valves is a main form of lymphatic dysfunction, known to result in severe edematous phenotypes; however, the extent of lymphatic valve deficiency in secondary lymphedema, including obesity-induced lymphedema, remains unknown. Therefore, the aims of the present study were 1) to determine whether western diet-induced obesity results in lymphatic valve dysfunction, and 2) to determine whether lymphatic valve dysfunction in western diet-induced obesity results from the diet itself, or as a consequence of the metabolic alterations induced by the diet. First, we quantitatively assessed and compared valve function in isolated popliteal and mesenteric collecting lymphatic vessels from control and western diet-induced obese C57BL/6J (WT) mice. Feeding a western diet for 14 weeks induced obesity and elevated plasma glucose and cholesterol levels when compared to controls. The function of lymphatic valves in popliteal lymphatics was not affected by diet-induced obesity; however, significant back-leak of pressure was observed in mesenteric lymphatic valves. Dysfunctional, leaky valves from obese animals also required significantly higher adverse pressure to trigger valve closure. Importantly, when subjected to treatment with a western diet, globally deficient PAI-1 mice were significantly protected against metabolic dysfunction and displayed fully functional, competent mesenteric lymphatic valves. In conclusion, our findings show for the first time that, in association with the metabolic alterations induced by the western diet, lymphatic valve dysfunction can be a critical component of obesity-induced lymphedema.
Collapse
|
14
|
Davis MJ, Scallan JP, Castorena-Gonzalez JA, Kim HJ, Ying LH, Pin YK, Angeli V. Multiple aspects of lymphatic dysfunction in an ApoE -/- mouse model of hypercholesterolemia. Front Physiol 2022; 13:1098408. [PMID: 36685213 PMCID: PMC9852907 DOI: 10.3389/fphys.2022.1098408] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 12/19/2022] [Indexed: 01/07/2023] Open
Abstract
Introduction: Rodent models of cardiovascular disease have uncovered various types of lymphatic vessel dysfunction that occur in association with atherosclerosis, type II diabetes and obesity. Previously, we presented in vivo evidence for impaired lymphatic drainage in apolipoprotein E null (ApoE -/- ) mice fed a high fat diet (HFD). Whether this impairment relates to the dysfunction of collecting lymphatics remains an open question. The ApoE -/- mouse is a well-established model of cardiovascular disease, in which a diet rich in fat and cholesterol on an ApoE deficient background accelerates the development of hypercholesteremia, atherosclerotic plaques and inflammation of the skin and other tissues. Here, we investigated various aspects of lymphatic function using ex vivo tests of collecting lymphatic vessels from ApoE +/+ or ApoE -/- mice fed a HFD. Methods: Popliteal collectors were excised from either strain and studied under defined conditions in which we could quantify changes in lymphatic contractile strength, lymph pump output, secondary valve function, and collecting vessel permeability. Results: Our results show that all these aspects of lymphatic vessel function are altered in deleterious ways in this model of hypercholesterolemia. Discussion: These findings extend previous in vivo observations suggesting significant dysfunction of lymphatic endothelial cells and smooth muscle cells from collecting vessels in association with a HFD on an ApoE-deficient background. An implication of our study is that collecting vessel dysfunction in this context may negatively impact the removal of cholesterol by the lymphatic system from the skin and the arterial wall and thereby exacerbate the progression and/or severity of atherosclerosis and associated inflammation.
Collapse
Affiliation(s)
- Michael J Davis
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, United States
| | - Joshua P Scallan
- Department of Molecular Pharmacology, University of South Florida, Tampa, FL, United States
| | | | - Hae Jin Kim
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, United States
| | - Lim Hwee Ying
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore
| | - Yeo Kim Pin
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore.,Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Veronique Angeli
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, Department of Microbiology and Immunology, National University of Singapore, Singapore, Singapore.,Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| |
Collapse
|
15
|
Scallan JP, Knauer LA, Hou H, Castorena-Gonzalez JA, Davis MJ, Yang Y. Foxo1 deletion promotes the growth of new lymphatic valves. J Clin Invest 2021; 131:e142341. [PMID: 34263740 DOI: 10.1172/jci142341] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 06/03/2021] [Indexed: 11/17/2022] Open
Abstract
Patients with congenital lymphedema suffer from tissue swelling in part due to mutations in genes regulating lymphatic valve development. Lymphatic valve leaflets grow and are maintained throughout life in response to oscillatory shear stress (OSS), which regulates gene transcription in lymphatic endothelial cells (LECs). Here, we identified the first transcription factor, Foxo1, that repressed lymphatic valve formation by inhibiting the expression of valve-forming genes. We showed that both embryonic and postnatal ablation of Foxo1 in LECs induced additional valve formation in postnatal and adult mice in multiple tissues. Our quantitative analyses revealed that after deletion, the total number of valves in the mesentery was significantly (P < 0.01) increased in the Foxo1LEC-KO mice compared with Foxo1fl/fl controls. In addition, our quantitative real-time PCR (RT-PCR) data from cultured LECs showed that many valve-forming genes were significantly (P < 0.01) upregulated upon knockdown of FOXO1. To confirm our findings in vivo, rescue experiments showed that Foxc2+/- mice, a model of lymphedema-distichiasis, had 50% fewer lymphatic valves and that the remaining valves exhibited backleak. Both valve number and function were completely restored to control levels upon Foxo1 deletion. These findings established FOXO1 as a clinically relevant target to stimulate de novo lymphatic valve formation and rescue defective valves in congenital lymphedema.
Collapse
Affiliation(s)
- Joshua P Scallan
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Luz A Knauer
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Huayan Hou
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | | | - Michael J Davis
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri, USA
| | - Ying Yang
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| |
Collapse
|
16
|
Bertram CD, Ikhimwin BO, Macaskill C. Modeling flow in embryonic lymphatic vasculature: what is its role in valve development? MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2021; 18:1406-1424. [PMID: 33757191 DOI: 10.3934/mbe.2021073] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
A majority of lymphatic valves tend to form in proximity to vessel junctions, and it is often proposed that disturbed flow at junctions creates oscillating shear stress that leads to accumulation of transcription factors which bring about valvogenesis at these sites. In images of networks of dorsal skin lymphatics from embryonic mice (day E16), we compared simulated fluid flow patterns and observed distributions of the transcription factor Prox1, which is implicated in valve formation. Because of creeping-flow conditions, flow across vessel junctions was not 'disturbed', and within a given vessel, shear stress varied inversely with local conduit width. Prox1 concentration was indeed localised to vessel end-regions, but over three networks was not consistently correlated with the vessel normalised-distance distribution of either fluid shear stress or shear-stress axial gradient. These findings do not support the presently accepted mechanism for the role of flow in valve localisation.
Collapse
Affiliation(s)
- Christopher D Bertram
- School of Mathematics and Statistics, University of Sydney, New South Wales 2006, Australia
| | - Bernard O Ikhimwin
- School of Mathematics and Statistics, University of Sydney, New South Wales 2006, Australia
| | - Charlie Macaskill
- School of Mathematics and Statistics, University of Sydney, New South Wales 2006, Australia
| |
Collapse
|
17
|
Blood and lymphatic systems are segregated by the FLCN tumor suppressor. Nat Commun 2020; 11:6314. [PMID: 33298956 PMCID: PMC7725783 DOI: 10.1038/s41467-020-20156-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 11/16/2020] [Indexed: 12/29/2022] Open
Abstract
Blood and lymphatic vessels structurally bear a strong resemblance but never share a lumen, thus maintaining their distinct functions. Although lymphatic vessels initially arise from embryonic veins, the molecular mechanism that maintains separation of these two systems has not been elucidated. Here, we show that genetic deficiency of Folliculin, a tumor suppressor, leads to misconnection of blood and lymphatic vessels in mice and humans. Absence of Folliculin results in the appearance of lymphatic-biased venous endothelial cells caused by ectopic expression of Prox1, a master transcription factor for lymphatic specification. Mechanistically, this phenotype is ascribed to nuclear translocation of the basic helix-loop-helix transcription factor Transcription Factor E3 (TFE3), binding to a regulatory element of Prox1, thereby enhancing its venous expression. Overall, these data demonstrate that Folliculin acts as a gatekeeper that maintains separation of blood and lymphatic vessels by limiting the plasticity of committed endothelial cells. Blood and lymphatic vessels bear a strong resemblance but do not share a lumen, thus maintaining their distinct functions. Here, the authors describe that Folliculin, a tumor suppressor, prevents the fusion of these vessels during development by limiting the plasticity of venous and lymphatic endothelial cells.
Collapse
|
18
|
Norden PR, Sabine A, Wang Y, Demir CS, Liu T, Petrova TV, Kume T. Shear stimulation of FOXC1 and FOXC2 differentially regulates cytoskeletal activity during lymphatic valve maturation. eLife 2020; 9:53814. [PMID: 32510325 PMCID: PMC7302880 DOI: 10.7554/elife.53814] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 06/06/2020] [Indexed: 12/14/2022] Open
Abstract
Mutations in the transcription factor FOXC2 are predominately associated with lymphedema. Herein, we demonstrate a key role for related factor FOXC1, in addition to FOXC2, in regulating cytoskeletal activity in lymphatic valves. FOXC1 is induced by laminar, but not oscillatory, shear and inducible, endothelial-specific deletion impaired postnatal lymphatic valve maturation in mice. However, deletion of Foxc2 induced valve degeneration, which is exacerbated in Foxc1; Foxc2 mutants. FOXC1 knockdown (KD) in human lymphatic endothelial cells increased focal adhesions and actin stress fibers whereas FOXC2-KD increased focal adherens and disrupted cell junctions, mediated by increased ROCK activation. ROCK inhibition rescued cytoskeletal or junctional integrity changes induced by inactivation of FOXC1 and FOXC2 invitro and vivo respectively, but only ameliorated valve degeneration in Foxc2 mutants. These results identify both FOXC1 and FOXC2 as mediators of mechanotransduction in the postnatal lymphatic vasculature and posit cytoskeletal signaling as a therapeutic target in lymphatic pathologies.
Collapse
Affiliation(s)
- Pieter R Norden
- Feinberg Cardiovascular and Renal Research Institute, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, United States
| | - Amélie Sabine
- Department of Oncology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Epalinges, Switzerland
| | - Ying Wang
- Department of Biochemistry and Molecular Biology, College of Medicine and Science, Mayo Clinic, Jacksonville, United States
| | - Cansaran Saygili Demir
- Department of Oncology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Epalinges, Switzerland
| | - Ting Liu
- Feinberg Cardiovascular and Renal Research Institute, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, United States
| | - Tatiana V Petrova
- Department of Oncology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Epalinges, Switzerland
| | - Tsutomu Kume
- Feinberg Cardiovascular and Renal Research Institute, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, United States
| |
Collapse
|
19
|
Castorena-Gonzalez JA, Srinivasan RS, King PD, Simon AM, Davis MJ. Simplified method to quantify valve back-leak uncovers severe mesenteric lymphatic valve dysfunction in mice deficient in connexins 43 and 37. J Physiol 2020; 598:2297-2310. [PMID: 32267537 PMCID: PMC8170716 DOI: 10.1113/jp279472] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 03/25/2020] [Indexed: 12/16/2022] Open
Abstract
KEY POINTS Lymphatic valve defects are one of the major causes of lymph transport dysfunction; however, there are no accessible methods for quantitatively assessing valve function. This report describes a novel technique for quantifying lymphatic valve back-leak. Postnatal endothelial-specific deletion of connexin 43 (Cx43) in connexin 37 null (Cx37-/- ) mice results in rapid regression of valve leaflets and severe valve dysfunction. This method can also be used for assessing the function of venous and lymphatic valves from various species, including humans. ABSTRACT The lymphatic system relies on robust, spontaneous contractions of collecting lymphatic vessels and one-way secondary lymphatic valves to efficiently move lymph forward. Secondary valves prevent reflux and allow for the generation of propulsive pressure during each contraction cycle. Lymphatic valve defects are one of the major causes of lymph transport dysfunction. Genetic mutations in multiple genes have been associated with the development of primary lymphoedema in humans; and many of the same mutations in mice result in valve defects that subsequently lead to chylous ascites or chylothorax. At present the only experimental technique for the quantitative assessment of lymphatic valve function utilizes the servo-null micropressure system, which is highly accurate and precise, but relatively inaccessible and difficult to use. We developed a novel, simplified alternative method for quantifying valve function and determining the degree of pressure back-leak through an intact valve in pressurized, single-valve segments of isolated lymphatic vessels. With this diameter-based method, the competence of each lymphatic valve is challenged over a physiological range of pressures (e.g. 0.5-10cmH2 O) and pressure back-leak is extrapolated from calibrated, pressure-driven changes in diameter upstream from the valve. Using mesenteric lymphatic vessels from C57BL/6J, Ub-CreERT2 ;Rasa1fx/fx , Foxc2Cre/+ , Lyve1-Cre;Cx43fx/fx , and Prox1-CreERT2 ;Cx43fx/fx ;Cx37-/- mice, we tested our method on lymphatic valves displaying a wide range of dysfunction, from fully competent to completely incompetent. Our results were validated by simultaneous direct measurement of pressure back-leak using a servo-null micropressure system. Our diameter-based technique can be used to quantify valve function in isolated lymphatic valves from a variety of species. This method also revealed that haplodeficiency in Foxc2 (Foxc2Cre/+ ) is not sufficient to cause significant valve dysfunction; however, postnatal endothelial-specific deletion of Cx43 in Cx37-/- mice results in rapid regression of valve leaflets and severe valve dysfunction.
Collapse
Affiliation(s)
- Jorge A Castorena-Gonzalez
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO, USA
| | - R Sathish Srinivasan
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Philip D King
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | | | - Michael J Davis
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO, USA
| |
Collapse
|
20
|
Hokkanen K, Tirronen A, Ylä-Herttuala S. Intestinal lymphatic vessels and their role in chylomicron absorption and lipid homeostasis. Curr Opin Lipidol 2019; 30:370-376. [PMID: 31361624 DOI: 10.1097/mol.0000000000000626] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW In this review, we describe novel findings related to intestinal lipid transport in lymphatic vessels. RECENT FINDINGS Studies have shown that chylomicron entry to lacteals and lymph movement in intestinal lymphatic capillaries is an active process. Regulators of this intestinal chylomicron transport include among others the autonomous nervous system, transcription factors like PLAGL2, and molecular regulators, such as VEGF-A/Nrp1/VEGFR1, VEGF-C/VEGFR3, DLL4, CALCRL and GLP-2. Chylomicron transport in intestinal lymphatics is now emerging not only as an option for drug delivery but also as a new candidate for drug targeting in lipid-related disorders. SUMMARY Dysfunctions of lymphatic lipid transport can result in conditions such as dyslipidaemia. Intestinal lymphatics also provide several potential therapeutic possibilities: molecular regulation of lacteal cell-to-cell junctioning and lymph flow could provide new ways of treating conditions like hyperlipidaemia and associated diseases, such as atherosclerosis and other cardiovascular diseases, obesity, diabetes and fatty-liver disease. The intestinal lymphatic system can also be employed to deliver lipid nanoparticles as drug carriers to the venous circulation for improved treatment outcome. These findings highlight the importance and need for research on the different players of intestinal lymphatics in dietary lipid handling and therapeutic applications.
Collapse
Affiliation(s)
- Krista Hokkanen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland
| | - Annakaisa Tirronen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland
| | - Seppo Ylä-Herttuala
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland
- Heart Center and Gene Therapy Unit, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
21
|
Liang Q, Zhang L, Wood RW, Ji RC, Boyce BF, Schwarz EM, Wang Y, Xing L. Avian Reticuloendotheliosis Viral Oncogene Related B Regulates Lymphatic Endothelial Cells during Vessel Maturation and Is Required for Lymphatic Vessel Function in Adult Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:2516-2530. [PMID: 31539516 DOI: 10.1016/j.ajpath.2019.08.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 08/05/2019] [Accepted: 08/22/2019] [Indexed: 12/16/2022]
Abstract
NF-κB signals through canonical transcription factor p65 (RelA)/p50 and noncanonical avian reticuloendotheliosis viral oncogene related B (RelB)/p52 pathways. The RelA/p50 is involved in basal and inflammatory lymphangiogenesis. However, the role of RelB/p52 in lymphatic vessel biology is unknown. Herein, we investigated changes in lymphatic vessels (LVs) in mice deficient in noncanonical NF-κB signaling and the function of RelB in lymphatic endothelial cells (LECs). LVs were examined in Relb-/-, p52-/-, or control mice, and the gene expression profiles in LECs with RelB knockdown. Relb-/-, but not p52-/-, mice exhibited multiple LV abnormalities. They include the following: i) increased capillary vessel diameter, ii) reduced smooth muscle cell (SMC) coverage of mature vessels, iii) leakage, and iv) loss of active and passive lymphatic flow. Relb-/- mature LVs had thinner vessel walls, more apoptotic LECs and SMCs, and fewer LEC junctions. RelB knockdown LECs had decreased growth, survival, and adhesion, and dysregulated signaling pathways involving these cellular events. These results suggest that Relb-/- mice have abnormal LVs, mainly in mature vessels with reduced SMC coverage, leakage, and loss of contractions. RelB knockdown in LECs leads to reduced growth, survival, and adhesion. RelB plays a vital role in LEC-mediated LV maturation and function.
Collapse
Affiliation(s)
- Qianqian Liang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China; Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York; Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Li Zhang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China; Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York; Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Ronald W Wood
- Department of Obstetrics and Gynecology, Urology, and Neurobiology and Anatomy, University of Rochester Medical Center, Rochester, New York
| | | | - Brendan F Boyce
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York; Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York
| | - Edward M Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York
| | - Yongjun Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China; Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China.
| | - Lianping Xing
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York; Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York.
| |
Collapse
|