1
|
Chen SD, Chu CY, Wang CB, Yang Y, Xu ZY, Qu YL, Man Y. Integrated-omics profiling unveils the disparities of host defense to ECM scaffolds during wound healing in aged individuals. Biomaterials 2024; 311:122685. [PMID: 38944969 DOI: 10.1016/j.biomaterials.2024.122685] [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: 02/05/2024] [Revised: 06/11/2024] [Accepted: 06/23/2024] [Indexed: 07/02/2024]
Abstract
Extracellular matrix (ECM) scaffold membranes have exhibited promising potential to better the outcomes of wound healing by creating a regenerative microenvironment around. However, when compared to the application in younger individuals, the performance of the same scaffold membrane in promoting re-epithelialization and collagen deposition was observed dissatisfying in aged mice. To comprehensively explore the mechanisms underlying this age-related disparity, we conducted the integrated analysis, combing single-cell RNA sequencing (scRNA-Seq) with spatial transcriptomics, and elucidated six functionally and spatially distinctive macrophage groups and lymphocytes surrounding the ECM scaffolds. Through intergroup comparative analysis and cell-cell communication, we characterized the dysfunction of Spp1+ macrophages in aged mice impeded the activation of the type Ⅱ immune response, thus inhibiting the repair ability of epidermal cells and fibroblasts around the ECM scaffolds. These findings contribute to a deeper understanding of biomaterial applications in varied physiological contexts, thereby paving the way for the development of precision-based biomaterials tailored specifically for aged individuals in future therapeutic strategies.
Collapse
Affiliation(s)
- Shuai-Dong Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Chen-Yu Chu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Chen-Bing Wang
- College & Hospital of Stomatology, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei, 230032, China
| | - Yang Yang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Zhao-Yu Xu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yi-Li Qu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yi Man
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
2
|
Matsuda T, Kono T, Taki Y, Sakuma I, Fujimoto M, Hashimoto N, Kawakami E, Fukuhara N, Nishioka H, Inoshita N, Yamada S, Nakamura Y, Horiguchi K, Miki T, Higuchi Y, Tanaka T. Deciphering craniopharyngioma subtypes: Single-cell analysis of tumor microenvironment and immune networks. iScience 2024; 27:111068. [PMID: 39483146 PMCID: PMC11525618 DOI: 10.1016/j.isci.2024.111068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 06/24/2024] [Accepted: 09/26/2024] [Indexed: 11/03/2024] Open
Abstract
Craniopharyngiomas, including adamantinomatous (ACP) and squamous papillary (PCP) types, are challenging to treat because of their proximity to crucial pituitary structures. This study aimed to characterize the cellular composition, tumor tissue diversity, and cell-cell interactions in ACPs and PCPs using single-cell RNA sequencing. Single-cell clustering revealed diverse cell types, further classified into developing epithelial, calcification, and immune response for ACP and developing epithelial, cell cycle, and immune response for PCP, based on gene expression patterns. Subclustering revealed the enrichment of classical M1 and M2 macrophages in ACP and PCP, respectively, with high expression of pro-inflammatory markers in classical M1 macrophages. The classical M1 and M2 macrophage ratio significantly correlated with the occurrence of diabetes insipidus and panhypopituitarism. Cell-cell interactions, particularly involving CD44-SPP, were identified between tumor cells. Thus, we developed a comprehensive cell atlas that elucidated the molecular characteristics and immune cell inter-networking in ACP and PCP tumor microenvironments.
Collapse
Affiliation(s)
- Tatsuma Matsuda
- Department of Neurological Surgery Chiba University Graduate School of Medicine, Chiba, Japan
- Department of Molecular Diagnosis, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Takashi Kono
- Department of Molecular Diagnosis, Graduate School of Medicine, Chiba University, Chiba, Japan
- Research Institute of Disaster Medicine, Chiba University, Chiba, Japan
| | - Yuki Taki
- Department of Molecular Diagnosis, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Ikki Sakuma
- Department of Molecular Diagnosis, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Masanori Fujimoto
- Department of Molecular Diagnosis, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Naoko Hashimoto
- Department of Molecular Diagnosis, Graduate School of Medicine, Chiba University, Chiba, Japan
- Research Institute of Disaster Medicine, Chiba University, Chiba, Japan
| | - Eiryo Kawakami
- Department of Aritificial Intelligence Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Noriaki Fukuhara
- Department of Hypothalamic and Pituitary Surgery, Toranomon Hospital, Tokyo, Japan
| | - Hiroshi Nishioka
- Department of Hypothalamic and Pituitary Surgery, Toranomon Hospital, Tokyo, Japan
| | - Naoko Inoshita
- Hypothalamic and Pituitary Center, Moriyama Memorial Hospital, Tokyo, Japan
| | - Shozo Yamada
- Hypothalamic and Pituitary Center, Moriyama Memorial Hospital, Tokyo, Japan
| | - Yasuhiro Nakamura
- Division of Pathology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Miyagi, Japan
| | - Kentaro Horiguchi
- Department of Neurological Surgery Chiba University Graduate School of Medicine, Chiba, Japan
| | - Takashi Miki
- Research Institute of Disaster Medicine, Chiba University, Chiba, Japan
- Department of Medical Physiology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Yoshinori Higuchi
- Department of Neurological Surgery Chiba University Graduate School of Medicine, Chiba, Japan
| | - Tomoaki Tanaka
- Department of Molecular Diagnosis, Graduate School of Medicine, Chiba University, Chiba, Japan
- Research Institute of Disaster Medicine, Chiba University, Chiba, Japan
| |
Collapse
|
3
|
Leifer VP, Fang F, Song L, Kim J, Papanikolaou JF, Smeeton J, Thomopoulos S. Single-cell RNA-sequencing analysis of immune and mesenchymal cell crosstalk in the developing enthesis. Sci Rep 2024; 14:26839. [PMID: 39500962 PMCID: PMC11538517 DOI: 10.1038/s41598-024-77958-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 10/28/2024] [Indexed: 11/08/2024] Open
Abstract
Autoimmunity underlies many painful disorders, such as enthesopathies, which localize to the enthesis. From infiltration of the synovium and axial skeleton by B cells, to disturbances in the ratio of M1/M2 enthesis macrophages, to CD8 + T cell mediated inflammation, autoimmune dysregulation is becoming increasingly well characterized in enthesopathies. Tissue resident B cells, macrophages, neutrophils, and T cells have also been localized in healthy human entheses. However, the potential developmental origins, presence, and role of immune cells (ICs) in enthesis development is not known. Here, we use single-cell RNA-sequencing analysis to describe IC subtypes present in the enthesis before, during, and after mineralization, and to infer regulatory interactions between ICs and mesenchymal cells (MCs). We report the presence of nine phenotypically distinct IC subtypes, including B cells, macrophages, neutrophils, and T cells. We find that specific IC subtypes may promote MC-proliferation and differentiation, and that MCs may regulate IC phenotype and autoimmunity. Our findings suggest that bidirectional regulatory interactions between ICs and MCs may be important to enthesis mineralization, and suggest that progenitor MCs have a unique ability to limit autoimmunity during development.
Collapse
Affiliation(s)
- Valia P Leifer
- Department of Orthopedic Surgery, Columbia University, New York, NY, 10032, USA
| | - Fei Fang
- Department Orthopedics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Lee Song
- Department of Orthopedic Surgery, Columbia University, New York, NY, 10032, USA
| | - Jieon Kim
- Department of Orthopedic Surgery, Columbia University, New York, NY, 10032, USA
| | - John F Papanikolaou
- Department of Orthopedic Surgery, Columbia University, New York, NY, 10032, USA
| | - Joanna Smeeton
- Department of Rehabilitation and Regenerative Medicine, Columbia Stem Cell Initiative, Columbia University, New York, NY, 10032, USA
- Department of Genetics and Development, Columbia Stem Cell Initiative, Columbia University, New York, NY, 10032, USA
| | - Stavros Thomopoulos
- Department of Orthopedic Surgery, Columbia University, New York, NY, 10032, USA.
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA.
| |
Collapse
|
4
|
Zhang H, Hasegawa Y, Suzuki M, Zhang T, Leitner DR, Jackson RP, Waldor MK. Mouse enteric neurons control intestinal plasmacytoid dendritic cell function via serotonin-HTR7 signaling. Nat Commun 2024; 15:9237. [PMID: 39455564 PMCID: PMC11511829 DOI: 10.1038/s41467-024-53545-2] [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: 03/19/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024] Open
Abstract
Serotonergic neurons in the central nervous system control behavior and mood, but knowledge of the roles of serotonergic circuits in the regulation of immune homeostasis is limited. Here, we employ mouse genetics to investigate the functions of enteric serotonergic neurons in the control of immune responses and find that these circuits regulate IgA induction and boost host defense against oral, but not systemic Salmonella Typhimurium infection. Enteric serotonergic neurons promote gut-homing, retention and activation of intestinal plasmacytoid dendritic cells (pDC). Mechanistically, this neuro-immune crosstalk is achieved through a serotonin-5-HT receptor 7 (HTR7) signaling axis that ultimately facilitates the pDC-mediated differentiation of IgA+ B cells from IgD+ precursors in the gut. Single-cell RNA-seq data further reveal novel patterns of bidirectional communication between specific subsets of enteric neurons and lamina propria DC. Our findings thus reveal a close interplay between enteric serotonergic neurons and gut immune homeostasis that enhances mucosal defense.
Collapse
Affiliation(s)
- Hailong Zhang
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, 02115, USA
- Howard Hughes Medical Institute, Boston, MA, 02115, USA
| | - Yuko Hasegawa
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, 02115, USA
- Howard Hughes Medical Institute, Boston, MA, 02115, USA
| | - Masataka Suzuki
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, 02115, USA
- Howard Hughes Medical Institute, Boston, MA, 02115, USA
| | - Ting Zhang
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, 02115, USA
- Howard Hughes Medical Institute, Boston, MA, 02115, USA
| | - Deborah R Leitner
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, 02115, USA
- Howard Hughes Medical Institute, Boston, MA, 02115, USA
| | - Ruaidhrí P Jackson
- Department of Immunology, Harvard Medical School, Boston, MA, 02115, USA
| | - Matthew K Waldor
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, 02115, USA.
- Department of Microbiology, Harvard Medical School, Boston, MA, 02115, USA.
- Howard Hughes Medical Institute, Boston, MA, 02115, USA.
| |
Collapse
|
5
|
Wang G, Zhang E, Chen A, Meng D. Single-cell RNA-seq analysis revealed the stemness of a specific cluster of B cells in acute lymphoblastic leukemia progression. PeerJ 2024; 12:e18296. [PMID: 39465162 PMCID: PMC11505884 DOI: 10.7717/peerj.18296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 09/22/2024] [Indexed: 10/29/2024] Open
Abstract
Background Childhood acute lymphoblastic leukemia (ALL) is a common pediatric cancer. The heterogeneous characterization of B cells in ALL progression poses new challenges to researchers. We used single-cell sequencing to explore the critical role of B cells in regulating the ALL immune microenvironment. Method We collected the single cell (sc) RNA-seq data of ALL and health sample from the gene expression omnibus (GEO) database, the "Seurat" and "harmony" R package was used for quality control and scRNA-seq analysis, in which the CellMarker2.0 database was used for cell type annotation. Subsequently, the FindAllMarkers function was used to identify the differentially expressed genes (DEGs) among various cell types and the DAVID database was applied for the biological process of DEGs. Then, the "inferCNV" package was used for copy number variation, regulons and cell communication were performed by SCENIC tool and CellChat package. The role of the target gene in regulating ALL progression was assessed using RT-qPCR, Transwell and scratch healing assays. Results We identified nine mainly cell clusters after scRNA-seq analysis, in which the B cells had higher infiltration proportion in the ALL samples and were sub-clustered into five cell sub-groups. The B cells 1 is closely associated with cell proliferation and stemness (TNFAIP3 and KDM5B), and the significant CNV of amplification occurred on chr6 and chr21 that supported stemness of B cells1. RXRB is a key transcription factor mediated the proliferation of B cells 1, which in turn suppressed hematopoietic stem cells (HSCs) proliferation and promoted cytotoxic NK/T cells activation through diverse cell communication ways. One of the key regulators of B cells is MYC, which promotes the migration and invasive ability of cell line leukemia cell lines. Conclusion This study reveals the stemness characteristics of B cells and their critical role in ALL progression, a finding that provides new potential directions for the development of targeted therapies against ALL.
Collapse
Affiliation(s)
- Guifang Wang
- Department of Pediatric Medicine, The Second Affiliated Hospital of Shandong First Medical University, Tai’an, Shandong, China
| | - Ensheng Zhang
- Department of Pediatric Hematology, Shandong Maternal and Child Health Hospital, Jinan, Shandong, China
| | - An Chen
- Department of Otolaryngology, The Second Affiliated Hospital of Shandong First Medical University, Tai’an, Shandong, China
| | - Dachuan Meng
- Department of Pediatric Hematology, Shandong Maternal and Child Health Hospital, Jinan, Shandong, China
| |
Collapse
|
6
|
Wu J, Lei JH, Li M, Zhang A, Li Y, Liang X, de Souza SC, Yuan Z, Wang C, Chen G, Liu TM, Deng CX, Tang Z, Qu S. Carbon Dots Crosslinked Egg White Hydrogel for Tissue Engineering. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2404702. [PMID: 39303206 DOI: 10.1002/advs.202404702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 09/01/2024] [Indexed: 09/22/2024]
Abstract
Egg white (EW)-derived hydrogels hold promise as biomaterials for in vitro cell culture due to their ability to mimic the extracellular matrix. However, their highly cross-linked structures restrict their potential for in vivo applications, as they are unable to integrate dynamically with tissues before degradation. In this study, this limitation is addressed by introducing carbon dots (CDs) as cross-linking agents for EW in a dilute aqueous solution. The resulting CDs-crosslinked EW hydrogel (CEWH) exhibits tensile strength comparable to that of skin tissue and features a large pore structure that promotes cell infiltration. Subcutaneous implantation of CEWH demonstrated excellent integration with surrounding tissue and a degradation rate aligned with the hair follicles (HFs) regeneration cycle. This allows the long-term regeneration and establishment of an M2 macrophage-dominated immune microenvironment, which in turn promotes the re-entry of HFs into the anagen phase from the telogen phase. Additionally, CEWH demonstrated potential as a wound dressing material. Overall, this study paves the way for utilizing EW as a versatile biomaterial for tissue engineering.
Collapse
Affiliation(s)
- Jun Wu
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Macau, 999078, P. R. China
- Department of Physics and Chemistry, Faculty of Science and Technology University of Macau, Macao, 999078, P. R. China
- MOE Frontier Science Centre for Precision Oncology University of Macau, Macao, 999078, P. R. China
| | - Josh Haipeng Lei
- MOE Frontier Science Centre for Precision Oncology University of Macau, Macao, 999078, P. R. China
- Faculty of Health Sciences, University of Macau, Macau, 999078, P. R. China
| | - Moxin Li
- MOE Frontier Science Centre for Precision Oncology University of Macau, Macao, 999078, P. R. China
- Faculty of Health Sciences, University of Macau, Macau, 999078, P. R. China
| | - Aiping Zhang
- MOE Frontier Science Centre for Precision Oncology University of Macau, Macao, 999078, P. R. China
- Faculty of Health Sciences, University of Macau, Macau, 999078, P. R. China
| | - Yuan Li
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Macau, 999078, P. R. China
- Department of Physics and Chemistry, Faculty of Science and Technology University of Macau, Macao, 999078, P. R. China
- MOE Frontier Science Centre for Precision Oncology University of Macau, Macao, 999078, P. R. China
| | - Xiao Liang
- MOE Frontier Science Centre for Precision Oncology University of Macau, Macao, 999078, P. R. China
- Faculty of Health Sciences, University of Macau, Macau, 999078, P. R. China
| | - Senio Campos de Souza
- Institute of Chinese Medical Sciences & State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macau, 999078, P. R. China
| | - Zhen Yuan
- MOE Frontier Science Centre for Precision Oncology University of Macau, Macao, 999078, P. R. China
- Faculty of Health Sciences, University of Macau, Macau, 999078, P. R. China
| | - Chunming Wang
- Institute of Chinese Medical Sciences & State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macau, 999078, P. R. China
| | - Guokai Chen
- MOE Frontier Science Centre for Precision Oncology University of Macau, Macao, 999078, P. R. China
- Faculty of Health Sciences, University of Macau, Macau, 999078, P. R. China
| | - Tzu-Ming Liu
- MOE Frontier Science Centre for Precision Oncology University of Macau, Macao, 999078, P. R. China
- Faculty of Health Sciences, University of Macau, Macau, 999078, P. R. China
| | - Chu-Xia Deng
- MOE Frontier Science Centre for Precision Oncology University of Macau, Macao, 999078, P. R. China
- Faculty of Health Sciences, University of Macau, Macau, 999078, P. R. China
| | - Zikang Tang
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Macau, 999078, P. R. China
- Department of Physics and Chemistry, Faculty of Science and Technology University of Macau, Macao, 999078, P. R. China
- MOE Frontier Science Centre for Precision Oncology University of Macau, Macao, 999078, P. R. China
| | - Songnan Qu
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Macau, 999078, P. R. China
- Department of Physics and Chemistry, Faculty of Science and Technology University of Macau, Macao, 999078, P. R. China
- MOE Frontier Science Centre for Precision Oncology University of Macau, Macao, 999078, P. R. China
| |
Collapse
|
7
|
Kousa AI, Jahn L, Zhao K, Flores AE, Acenas D, Lederer E, Argyropoulos KV, Lemarquis AL, Granadier D, Cooper K, D'Andrea M, Sheridan JM, Tsai J, Sikkema L, Lazrak A, Nichols K, Lee N, Ghale R, Malard F, Andrlova H, Velardi E, Youssef S, Burgos da Silva M, Docampo M, Sharma R, Mazutis L, Wimmer VC, Rogers KL, DeWolf S, Gipson B, Gomes ALC, Setty M, Pe'er D, Hale L, Manley NR, Gray DHD, van den Brink MRM, Dudakov JA. Age-related epithelial defects limit thymic function and regeneration. Nat Immunol 2024; 25:1593-1606. [PMID: 39112630 PMCID: PMC11362016 DOI: 10.1038/s41590-024-01915-9] [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: 05/26/2023] [Accepted: 07/03/2024] [Indexed: 09/01/2024]
Abstract
The thymus is essential for establishing adaptive immunity yet undergoes age-related involution that leads to compromised immune responsiveness. The thymus is also extremely sensitive to acute insult and although capable of regeneration, this capacity declines with age for unknown reasons. We applied single-cell and spatial transcriptomics, lineage-tracing and advanced imaging to define age-related changes in nonhematopoietic stromal cells and discovered the emergence of two atypical thymic epithelial cell (TEC) states. These age-associated TECs (aaTECs) formed high-density peri-medullary epithelial clusters that were devoid of thymocytes; an accretion of nonproductive thymic tissue that worsened with age, exhibited features of epithelial-to-mesenchymal transition and was associated with downregulation of FOXN1. Interaction analysis revealed that the emergence of aaTECs drew tonic signals from other functional TEC populations at baseline acting as a sink for TEC growth factors. Following acute injury, aaTECs expanded substantially, further perturbing trophic regeneration pathways and correlating with defective repair of the involuted thymus. These findings therefore define a unique feature of thymic involution linked to immune aging and could have implications for developing immune-boosting therapies in older individuals.
Collapse
Grants
- T32-GM007270 U.S. Department of Health & Human Services | NIH | National Institute of General Medical Sciences (NIGMS)
- 1187367 Department of Health | National Health and Medical Research Council (NHMRC)
- R01 CA228308 NCI NIH HHS
- 1158024 Department of Health | National Health and Medical Research Council (NHMRC)
- R01-HL145276 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01-HL147584 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01-HL165673 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HL123340 NHLBI NIH HHS
- R01 HL145276 NHLBI NIH HHS
- R01-CA228308 U.S. Department of Health & Human Services | NIH | National Cancer Institute (NCI)
- T32 GM007103 NIGMS NIH HHS
- P30 CA015704 NCI NIH HHS
- P01 CA023766 NCI NIH HHS
- R01 HL165673 NHLBI NIH HHS
- R01 HL147584 NHLBI NIH HHS
- P01-AG052359 U.S. Department of Health & Human Services | NIH | National Institute on Aging (U.S. National Institute on Aging)
- P30-CA015704 U.S. Department of Health & Human Services | NIH | National Cancer Institute (NCI)
- 1090236 Department of Health | National Health and Medical Research Council (NHMRC)
- P30 CA008748 NCI NIH HHS
- P01 AG052359 NIA NIH HHS
- T32 GM007270 NIGMS NIH HHS
- 1102104 Cancer Council Victoria
- 1078763 Department of Health | National Health and Medical Research Council (NHMRC)
- 1146518 Cancer Council Victoria
- U01-AI70035 U.S. Department of Health & Human Services | NIH | National Institute of Allergy and Infectious Diseases (NIAID)
- R35 HL171556 NHLBI NIH HHS
- ALTF-431-2017 European Molecular Biology Organization (EMBO)
- R01 CA228358 NCI NIH HHS
- F30-HL165761 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01-HL123340 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R35-HL-171556 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- 1121325 Department of Health | National Health and Medical Research Council (NHMRC)
- F30 HL165761 NHLBI NIH HHS
- U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- U.S. Department of Health & Human Services | NIH | National Institute of Allergy and Infectious Diseases (NIAID)
- U.S. Department of Health & Human Services | NIH | National Cancer Institute (NCI)
- U.S. Department of Health & Human Services | NIH | National Institute on Aging (U.S. National Institute on Aging)
- U.S. Department of Health & Human Services | NIH | National Institute of General Medical Sciences (NIGMS)
Collapse
Affiliation(s)
- Anastasia I Kousa
- Program in Immunology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Translational Science and Therapeutics Division, and Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Center, Seattle, WA, USA
- City of Hope Los Angeles and National Medical Center, Duarte, CA, USA
| | - Lorenz Jahn
- Program in Immunology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kelin Zhao
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Angel E Flores
- Department of Genetics, University of Georgia, Athens, GA, USA
| | - Dante Acenas
- Translational Science and Therapeutics Division, and Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Immunology, University of Washington, Seattle, WA, USA
| | - Emma Lederer
- Translational Science and Therapeutics Division, and Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Immunology, University of Washington, Seattle, WA, USA
| | - Kimon V Argyropoulos
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andri L Lemarquis
- Program in Immunology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- City of Hope Los Angeles and National Medical Center, Duarte, CA, USA
| | - David Granadier
- Translational Science and Therapeutics Division, and Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Kirsten Cooper
- Translational Science and Therapeutics Division, and Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Michael D'Andrea
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Julie M Sheridan
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Jennifer Tsai
- Program in Immunology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lisa Sikkema
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Institute of Computational Biology, Helmholtz Center Munich, Munich, Germany
| | - Amina Lazrak
- Program in Immunology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Katherine Nichols
- Program in Immunology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nichole Lee
- Program in Immunology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Romina Ghale
- Program in Immunology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Florent Malard
- Program in Immunology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Sorbonne Université, Centre de Recherche Saint-Antoine INSERM UMRs938, Service d'Hématologie Clinique et de Thérapie Cellulaire, Hôpital Saint Antoine, AP-HP, Paris, France
| | - Hana Andrlova
- Program in Immunology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Enrico Velardi
- Division of Pediatric Hematology and Oncology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Salma Youssef
- Program in Immunology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Melissa Docampo
- Program in Immunology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Roshan Sharma
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Linas Mazutis
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Verena C Wimmer
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Kelly L Rogers
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Susan DeWolf
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Brianna Gipson
- Program in Immunology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Antonio L C Gomes
- Program in Immunology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Manu Setty
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Basic Sciences Division & Translational Data Science Integrated Research Center, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Dana Pe'er
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Laura Hale
- Human Vaccine Institute, Duke University, Durham, NC, USA
| | - Nancy R Manley
- Department of Genetics, University of Georgia, Athens, GA, USA
- School of Life Sciences, Arizona State University, Phoenix, AZ, USA
| | - Daniel H D Gray
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia.
| | - Marcel R M van den Brink
- Program in Immunology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- City of Hope Los Angeles and National Medical Center, Duarte, CA, USA.
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Jarrod A Dudakov
- Translational Science and Therapeutics Division, and Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Center, Seattle, WA, USA.
- Department of Immunology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
8
|
Gu W, Eke C, Gonzalez Santiago E, Olaloye O, Konnikova L. Single-cell atlas of the small intestine throughout the human lifespan demonstrates unique features of fetal immune cells. Mucosal Immunol 2024; 17:599-617. [PMID: 38555026 PMCID: PMC11384551 DOI: 10.1016/j.mucimm.2024.03.011] [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: 11/02/2023] [Revised: 03/15/2024] [Accepted: 03/23/2024] [Indexed: 04/02/2024]
Abstract
Proper development of mucosal immunity is critical for human health. Over the past decade, it has become evident that in humans, this process begins in utero. However, there are limited data on the unique features and functions of fetal mucosal immune cells. To address this gap, we integrated several single-cell ribonucleic acid sequencing datasets of the human small intestine (SI) to create an SI transcriptional atlas throughout the human life span, ranging from the first trimester to adulthood, with a focus on immune cells. Fetal SI displayed a complex immune landscape comprising innate and adaptive immune cells that exhibited distinct transcriptional programs from postnatal samples, especially compared with pediatric and adult samples. We identified shifts in myeloid populations across gestation and progression of memory T-cell states throughout the human lifespan. In particular, there was a marked shift of memory T cells from those with stem-like properties in the fetal samples to fully differentiated cells with a high expression of activation and effector function genes in adult samples, with neonatal samples containing both features. Finally, we demonstrate that the SI developmental atlas can be used to elucidate improper trajectories linked to mucosal diseases by implicating developmental abnormalities underlying necrotizing enterocolitis, a severe intestinal complication of prematurity. Collectively, our data provide valuable resources and important insights into intestinal immunity that will facilitate regenerative medicine and disease understanding.
Collapse
Affiliation(s)
- Weihong Gu
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| | - Chino Eke
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| | | | - Oluwabunmi Olaloye
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| | - Liza Konnikova
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA; Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA; Department of Obstetrics, Gynecology and Reproductive Science, Yale University School of Medicine, New Haven, CT, USA; Program in Translational Biomedicine, Yale University School of Medicine, New Haven, CT, USA; Program in Human Translational Immunology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
9
|
von Roemeling CA, Patel JA, Carpenter SL, Yegorov O, Yang C, Bhatia A, Doonan BP, Russell R, Trivedi VS, Klippel K, Ryu DH, Grippin A, Futch HS, Ran Y, Hoang-Minh LB, Weidert FL, Golde TE, Mitchell DA. Adeno-associated virus delivered CXCL9 sensitizes glioblastoma to anti-PD-1 immune checkpoint blockade. Nat Commun 2024; 15:5871. [PMID: 38997283 PMCID: PMC11245621 DOI: 10.1038/s41467-024-49989-1] [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: 11/07/2023] [Accepted: 06/27/2024] [Indexed: 07/14/2024] Open
Abstract
There are numerous mechanisms by which glioblastoma cells evade immunological detection, underscoring the need for strategic combinatorial treatments to achieve appreciable therapeutic effects. However, developing combination therapies is difficult due to dose-limiting toxicities, blood-brain-barrier, and suppressive tumor microenvironment. Glioblastoma is notoriously devoid of lymphocytes driven in part by a paucity of lymphocyte trafficking factors necessary to prompt their recruitment and activation. Herein, we develop a recombinant adeno-associated virus (AAV) gene therapy that enables focal and stable reconstitution of the tumor microenvironment with C-X-C motif ligand 9 (CXCL9), a powerful call-and-receive chemokine for lymphocytes. By manipulating local chemokine directional guidance, AAV-CXCL9 increases tumor infiltration by cytotoxic lymphocytes, sensitizing glioblastoma to anti-PD-1 immune checkpoint blockade in female preclinical tumor models. These effects are accompanied by immunologic signatures evocative of an inflamed tumor microenvironment. These findings support AAV gene therapy as an adjuvant for reconditioning glioblastoma immunogenicity given its safety profile, tropism, modularity, and off-the-shelf capability.
Collapse
Affiliation(s)
- Christina A von Roemeling
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA.
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA.
| | - Jeet A Patel
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA
| | - Savannah L Carpenter
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA
| | - Oleg Yegorov
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA
| | - Changlin Yang
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA
| | - Alisha Bhatia
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA
| | - Bently P Doonan
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA
- Department of Medicine, Hematology and Oncology, University of Florida, Gainesville, FL, USA
| | - Rylynn Russell
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA
| | - Vrunda S Trivedi
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA
| | - Kelena Klippel
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA
| | - Daniel H Ryu
- Goizueta Brain Health Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Adam Grippin
- Department of Radiation Oncology, MD Anderson Cancer Center, The University of Texas, Houston, TX, USA
| | - Hunter S Futch
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Yong Ran
- Goizueta Brain Health Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Lan B Hoang-Minh
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA
| | - Frances L Weidert
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA
| | - Todd E Golde
- Goizueta Brain Health Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Duane A Mitchell
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA.
- Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
10
|
Jin M, Ma Z, Zhang H, Papetti AV, Dang R, Stillitano AC, Goldman SA, Jiang P. Co-Transplantation-Based Human-Mouse Chimeric Brain Models to Study Human Glial-Glial and Glial-Neuronal Interactions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.03.601990. [PMID: 39005270 PMCID: PMC11244967 DOI: 10.1101/2024.07.03.601990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Human-mouse chimeric brain models, generated by transplanting human induced pluripotent stem cell (hiPSC)-derived neural cells, are valuable for studying the development and function of human neural cells in vivo. Understanding glial-glial and glial-neuronal interactions is essential for unraveling the complexities of brain function and developing treatments for neurological disorders. To explore these interactions between human neural cells within an intact brain environment, we employe a co-transplantation strategy involving the engraftment of hiPSC-derived neural progenitor cells along with primitive macrophage progenitors into the neonatal mouse brain. This approach creates human-mouse chimeric brains containing human microglia, macroglia (astroglia and oligodendroglia), and neurons. Using super-resolution imaging and 3D reconstruction techniques, we examine the dynamics between human neurons and glia, unveiling human microglia engulfing immature human neurons, microglia pruning synapses of human neurons, and significant interactions between human oligodendrocytes and neurons. Single-cell RNA sequencing analysis of the chimeric brain uncovers a close recapitulation of the human glial progenitor cell population, along with a dynamic stage in astroglial development that mirrors the processes found in the human brain. Furthermore, cell-cell communication analysis highlights significant neuronal-glial and glial-glial interactions, especially the interaction between adhesion molecules neurexins and neuroligins. This innovative co-transplantation model opens up new avenues for exploring the complex pathophysiological mechanisms underlying human neurological diseases. It holds particular promise for studying disorders where glial-neuronal interactions and non-cell-autonomous effects play crucial roles.
Collapse
Affiliation(s)
- Mengmeng Jin
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
- These authors contributed equally
| | - Ziyuan Ma
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
- These authors contributed equally
| | - Haiwei Zhang
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
- These authors contributed equally
| | - Ava V. Papetti
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Rui Dang
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | | | - Steven A. Goldman
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, USA
- Center for Translational Neuromedicine, University of Copenhagen Faculty of Health and Medical Sciences, Copenhagen, Denmark
| | - Peng Jiang
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
- Lead Contact
| |
Collapse
|
11
|
Bolívar S, Sanz E, Ovelleiro D, Zochodne DW, Udina E. Neuron-specific RNA-sequencing reveals different responses in peripheral neurons after nerve injury. eLife 2024; 12:RP91316. [PMID: 38742628 PMCID: PMC11093584 DOI: 10.7554/elife.91316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2024] Open
Abstract
Peripheral neurons are heterogeneous and functionally diverse, but all share the capability to switch to a pro-regenerative state after nerve injury. Despite the assumption that the injury response is similar among neuronal subtypes, functional recovery may differ. Understanding the distinct intrinsic regenerative properties between neurons may help to improve the quality of regeneration, prioritizing the growth of axon subpopulations to their targets. Here, we present a comparative analysis of regeneration across four key peripheral neuron populations: motoneurons, proprioceptors, cutaneous mechanoreceptors, and nociceptors. Using Cre/Ai9 mice that allow fluorescent labeling of neuronal subtypes, we found that nociceptors showed the greater regeneration after a sciatic crush, followed by motoneurons, mechanoreceptors, and, finally, proprioceptors. By breeding these Cre mice with Ribotag mice, we isolated specific translatomes and defined the regenerative response of these neuronal subtypes after axotomy. Only 20% of the regulated genes were common, revealing a diverse response to injury among neurons, which was also supported by the differential influence of neurotrophins among neuron subtypes. Among differentially regulated genes, we proposed MED12 as a specific regulator of the regeneration of proprioceptors. Altogether, we demonstrate that the intrinsic regenerative capacity differs between peripheral neuron subtypes, opening the door to selectively modulate these responses.
Collapse
Affiliation(s)
- Sara Bolívar
- Institute of Neurosciences, and Department Cell Biology, Physiology and Immunology, Universitat Autònoma de BarcelonaBellaterraSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Instituto de Salud Carlos IIIMadridSpain
| | - Elisenda Sanz
- Institute of Neurosciences, and Department Cell Biology, Physiology and Immunology, Universitat Autònoma de BarcelonaBellaterraSpain
| | - David Ovelleiro
- Peripheral Nervous System, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital CampusBarcelonaSpain
| | - Douglas W Zochodne
- Division of Neurology, Department of Medicine and the Neuroscience and Mental Health Institute, University of AlbertaEdmontonCanada
| | - Esther Udina
- Institute of Neurosciences, and Department Cell Biology, Physiology and Immunology, Universitat Autònoma de BarcelonaBellaterraSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Instituto de Salud Carlos IIIMadridSpain
| |
Collapse
|
12
|
Tian Y, Guo J, Hua L, Jiang Y, Ge W, Zhang X, Cai D, Lu D, Wang B, Shen W, Sun Z, Han B. Mechanisms of imbalanced testicular homeostasis in infancy due to aberrant histone acetylation in undifferentiated spermatogonia under different concentrations of Di(2-ethylhexyl) phthalate (DEHP) exposure. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 347:123742. [PMID: 38460586 DOI: 10.1016/j.envpol.2024.123742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 03/03/2024] [Accepted: 03/06/2024] [Indexed: 03/11/2024]
Abstract
Di (2-ethylhexyl) phthalate (DEHP), identified as an endocrine-disrupting chemical, is associated with reproductive toxicity. This association is particularly noteworthy in newborns with incompletely developed metabolic functions, as exposure to DEHP can induce enduring damage to the reproductive system, potentially influencing adult reproductive health. In this study, we continuously administered 40 μg/kg and 80 μg/kg DEHP to postnatal day 5 (PD5) mice for ten days to simulate low and high doses of DEHP exposure during infancy. Utilizing single-cell RNA sequencing (scRNA-seq), our analysis revealed that varying concentrations of DEHP exposure during infancy induced distinct DNA damage response characteristics in testicular Undifferentiated spermatogonia (Undiff SPG). Specifically, DNA damage triggered mitochondrial dysfunction, leading to acetyl-CoA content alterations. Subsequently, this disruption caused aberrations in histone acetylation patterns, ultimately resulting in apoptosis of Undiff SPG in the 40 μg/kg DEHP group and autophagy in the 80 μg/kg DEHP group. Furthermore, we found that DEHP exposure impacts the development and functionality of Sertoli and Leydig cells through the focal adhesion and PPAR signaling pathways, respectively. We also revealed that Leydig cells regulate the metabolic environment of Undiff SPG via Ptn-Sdc4 and Mdk-Sdc4 after DEHP exposure. Finally, our study provided pioneering evidence that disruptions in testicular homeostasis induced by DEHP exposure during infancy endure into adulthood. In summary, this study elucidates the molecular mechanisms through which DEHP exposure during infancy influences the development of testicular cell populations.
Collapse
Affiliation(s)
- Yu Tian
- Department of Urology, Shenzhen University General Hospital, Shenzhen, China; College of Life Sciences, Qingdao Agricultural University, Qingdao, China
| | - Jiachen Guo
- College of Life Sciences, Qingdao Agricultural University, Qingdao, China
| | - Lei Hua
- School of Clinical Medicine, Henan University, Kaifeng, China
| | - Yinuo Jiang
- Department of Urology, Shenzhen University General Hospital, Shenzhen, China
| | - Wei Ge
- College of Life Sciences, Qingdao Agricultural University, Qingdao, China
| | - Xiaoyuan Zhang
- College of Life Sciences, Qingdao Agricultural University, Qingdao, China
| | - Diya Cai
- Department of Urology, Shenzhen University General Hospital, Shenzhen, China
| | - Dongliang Lu
- Department of Urology, Shenzhen University General Hospital, Shenzhen, China
| | - Bin Wang
- Department of Urology, Shenzhen University General Hospital, Shenzhen, China
| | - Wei Shen
- College of Life Sciences, Qingdao Agricultural University, Qingdao, China
| | - Zhongyi Sun
- Department of Urology, Shenzhen University General Hospital, Shenzhen, China
| | - Baoquan Han
- Department of Urology, Shenzhen University General Hospital, Shenzhen, China.
| |
Collapse
|
13
|
Trivedi V, Yang C, Klippel K, Yegorov O, von Roemeling C, Hoang-Minh L, Fenton G, Ogando-Rivas E, Castillo P, Moore G, Long-James K, Dyson K, Doonan B, Flores C, Mitchell DA. mRNA-based precision targeting of neoantigens and tumor-associated antigens in malignant brain tumors. Genome Med 2024; 16:17. [PMID: 38268001 PMCID: PMC10809449 DOI: 10.1186/s13073-024-01281-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: 04/14/2023] [Accepted: 01/02/2024] [Indexed: 01/26/2024] Open
Abstract
BACKGROUND Despite advancements in the successful use of immunotherapy in treating a variety of solid tumors, applications in treating brain tumors have lagged considerably. This is due, at least in part, to the lack of well-characterized antigens expressed within brain tumors that can mediate tumor rejection; the low mutational burden of these tumors that limits the abundance of targetable neoantigens; and the immunologically "cold" tumor microenvironment that hampers the generation of sustained and productive immunologic responses. The field of mRNA-based therapeutics has experienced a boon following the universal approval of COVID-19 mRNA vaccines. mRNA-based immunotherapeutics have also garnered widespread interest for their potential to revolutionize cancer treatment. In this study, we developed a novel and scalable approach for the production of personalized mRNA-based therapeutics that target multiple tumor rejection antigens in a single therapy for the treatment of refractory brain tumors. METHODS Tumor-specific neoantigens and aberrantly overexpressed tumor-associated antigens were identified for glioblastoma and medulloblastoma tumors using our cancer immunogenomics pipeline called Open Reading Frame Antigen Network (O.R.A.N). Personalized tumor antigen-specific mRNA vaccine was developed for each individual tumor model using selective gene capture and enrichment strategy. The immunogenicity and efficacy of the personalized mRNA vaccines was evaluated in combination with anti-PD-1 immune checkpoint blockade therapy or adoptive cellular therapy with ex vivo expanded tumor antigen-specific lymphocytes in highly aggressive murine GBM models. RESULTS Our results demonstrate the effectiveness of the antigen-specific mRNA vaccines in eliciting robust anti-tumor immune responses in GBM hosts. Our findings substantiate an increase in tumor-infiltrating lymphocytes characterized by enhanced effector function, both intratumorally and systemically, after antigen-specific mRNA-directed immunotherapy, resulting in a favorable shift in the tumor microenvironment from immunologically cold to hot. Capacity to generate personalized mRNA vaccines targeting human GBM antigens was also demonstrated. CONCLUSIONS We have established a personalized and customizable mRNA-therapeutic approach that effectively targets a plurality of tumor antigens and demonstrated potent anti-tumor response in preclinical brain tumor models. This platform mRNA technology uniquely addresses the challenge of tumor heterogeneity and low antigen burden, two key deficiencies in targeting the classically immunotherapy-resistant CNS malignancies, and possibly other cold tumor types.
Collapse
Affiliation(s)
- Vrunda Trivedi
- University of Florida, 1333 Center Drive, BSB B1-118, Gainesville, FL, 32610, USA
| | - Changlin Yang
- University of Florida, 1333 Center Drive, BSB B1-118, Gainesville, FL, 32610, USA
| | - Kelena Klippel
- University of Florida, 1333 Center Drive, BSB B1-118, Gainesville, FL, 32610, USA
| | - Oleg Yegorov
- University of Florida, 1333 Center Drive, BSB B1-118, Gainesville, FL, 32610, USA
| | | | - Lan Hoang-Minh
- University of Florida, 1333 Center Drive, BSB B1-118, Gainesville, FL, 32610, USA
| | - Graeme Fenton
- University of Florida, 1333 Center Drive, BSB B1-118, Gainesville, FL, 32610, USA
| | | | - Paul Castillo
- University of Florida, 1333 Center Drive, BSB B1-118, Gainesville, FL, 32610, USA
| | - Ginger Moore
- University of Florida, 1333 Center Drive, BSB B1-118, Gainesville, FL, 32610, USA
| | - Kaytora Long-James
- University of Florida, 1333 Center Drive, BSB B1-118, Gainesville, FL, 32610, USA
| | - Kyle Dyson
- University of Florida, 1333 Center Drive, BSB B1-118, Gainesville, FL, 32610, USA
| | - Bently Doonan
- University of Florida, 1333 Center Drive, BSB B1-118, Gainesville, FL, 32610, USA
| | - Catherine Flores
- University of Florida, 1333 Center Drive, BSB B1-118, Gainesville, FL, 32610, USA
| | - Duane A Mitchell
- University of Florida, 1333 Center Drive, BSB B1-118, Gainesville, FL, 32610, USA.
| |
Collapse
|
14
|
von Roemeling C, Yegorov O, Yang C, Klippel K, Russell R, Trivedi V, Bhatia A, Doonan B, Carpenter S, Ryu D, Grippen A, Futch H, Ran Y, Hoang-Minh L, Weidert F, Golde T, Mitchell D. CXCL9 recombinant adeno-associated virus (AAV) virotherapy sensitizes glioblastoma (GBM) to anti-PD-1 immune checkpoint blockade. RESEARCH SQUARE 2023:rs.3.rs-3463730. [PMID: 38014191 PMCID: PMC10680939 DOI: 10.21203/rs.3.rs-3463730/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
The promise of immunotherapy to induce long-term durable responses in conventionally treatment resistant tumors like glioblastoma (GBM) has given hope for patients with a dismal prognosis. Yet, few patients have demonstrated a significant survival benefit despite multiple clinical trials designed to invigorate immune recognition and tumor eradication. Insights gathered over the last two decades have revealed numerous mechanisms by which glioma cells resist conventional therapy and evade immunological detection, underscoring the need for strategic combinatorial treatments as necessary to achieve appreciable therapeutic effects. However, new combination therapies are inherently difficult to develop as a result of dose-limiting toxicities, the constraints of the blood-brain barrier, and the suppressive nature of the GBM tumor microenvironment (TME). GBM is notoriously devoid of lymphocytes driven in part by a paucity of lymphocyte trafficking factors necessary to prompt their recruitment, infiltration, and activation. We have developed a novel recombinant adeno-associated virus (AAV) gene therapy strategy that enables focal and stable reconstitution of the GBM TME with C-X-C motif ligand 9 (CXCL9), a powerful call-and-receive chemokine for cytotoxic T lymphocytes (CTLs). By precisely manipulating local chemokine directional guidance, AAV-CXCL9 increases tumor infiltration by CD8-postive cytotoxic lymphocytes, sensitizing GBM to anti-PD-1 immune checkpoint blockade (ICB). These effects are accompanied by immunologic signatures evocative of an inflamed and responsive TME. These findings support targeted AAV gene therapy as a promising adjuvant strategy for reconditioning GBM immunogenicity given its excellent safety profile, TME-tropism, modularity, and off-the-shelf capability, where focal delivery bypasses the constrains of the blood-brain barrier, further mitigating risks observed with high-dose systemic therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Frances Weidert
- Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida
| | | | | |
Collapse
|
15
|
Zhou Z, Pan Y, Zhou S, Wang S, Zhang D, Cao Y, Jiang X, Li J, Zhu L, Zhao L, Gu S, Lin G, Dong Z, Sun HX. Single-cell analysis reveals specific neuronal transition during mouse corticogenesis. Front Cell Dev Biol 2023; 11:1209320. [PMID: 38020907 PMCID: PMC10657809 DOI: 10.3389/fcell.2023.1209320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 10/24/2023] [Indexed: 12/01/2023] Open
Abstract
Background: Currently, the mechanism(s) underlying corticogenesis is still under characterization. Methods: We curated the most comprehensive single-cell RNA-seq (scRNA-seq) datasets from mouse and human fetal cortexes for data analysis and confirmed the findings with co-immunostaining experiments. Results: By analyzing the developmental trajectories with scRNA-seq datasets in mice, we identified a specific developmental sub-path contributed by a cell-population expressing both deep- and upper-layer neurons (DLNs and ULNs) specific markers, which occurred on E13.5 but was absent in adults. In this cell-population, the percentages of cells expressing DLN and ULN markers decreased and increased, respectively, during the development suggesting direct neuronal transition (namely D-T-U). Whilst genes significantly highly/uniquely expressed in D-T-U cell population were significantly enriched in PTN/MDK signaling pathways related to cell migration. Both findings were further confirmed by co-immunostaining with DLNs, ULNs and D-T-U specific markers across different timepoints. Furthermore, six genes (co-expressed with D-T-U specific markers in mice) showing a potential opposite temporal expression between human and mouse during fetal cortical development were associated with neuronal migration and cognitive functions. In adult prefrontal cortexes (PFC), D-T-U specific genes were expressed in neurons from different layers between humans and mice. Conclusion: Our study characterizes a specific cell population D-T-U showing direct DLNs to ULNs neuronal transition and migration during fetal cortical development in mice. It is potentially associated with the difference of cortical development in humans and mice.
Collapse
Affiliation(s)
- Ziheng Zhou
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Yueyang Pan
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Si Zhou
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Shuguang Wang
- Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Dengwei Zhang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Ye Cao
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Xiaosen Jiang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Jie Li
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Linnan Zhu
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, School of Life Sciences, Peking University, Beijing, China
| | - Lijian Zhao
- Medical Technology College, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Shen Gu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Ge Lin
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Zirui Dong
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Hai-Xi Sun
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
16
|
Qiu X, Guo Y, Liu M, Zhang B, Li J, Wei J, Li M. Single-cell RNA-sequencing analysis reveals enhanced non-canonical neurotrophic factor signaling in the subacute phase of traumatic brain injury. CNS Neurosci Ther 2023; 29:3446-3459. [PMID: 37269057 PMCID: PMC10580338 DOI: 10.1111/cns.14278] [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: 02/09/2023] [Revised: 04/25/2023] [Accepted: 05/14/2023] [Indexed: 06/04/2023] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) is a leading cause of long-term disability in young adults and induces complex neuropathological processes. Cellular autonomous and intercellular changes during the subacute phase contribute substantially to the neuropathology of TBI. However, the underlying mechanisms remain elusive. In this study, we explored the dysregulated cellular signaling during the subacute phase of TBI. METHODS Single-cell RNA-sequencing data (GSE160763) of TBI were analyzed to explore the cell-cell communication in the subacute phase of TBI. Upregulated neurotrophic factor signaling was validated in a mouse model of TBI. Primary cell cultures and cell lines were used as in vitro models to examine the potential mechanisms affecting signaling. RESULTS Single-cell RNA-sequencing analysis revealed that microglia and astrocytes were the most affected cells during the subacute phase of TBI. Cell-cell communication analysis demonstrated that signaling mediated by the non-canonical neurotrophic factors midkine (MDK), pleiotrophin (PTN), and prosaposin (PSAP) in the microglia/astrocytes was upregulated in the subacute phase of TBI. Time-course profiling showed that MDK, PTN, and PSAP expression was primarily upregulated in the subacute phase of TBI, and astrocytes were the major source of MDK and PTN after TBI. In vitro studies revealed that the expression of MDK, PTN, and PSAP in astrocytes was enhanced by activated microglia. Moreover, MDK and PTN promoted the proliferation of neural progenitors derived from human-induced pluripotent stem cells (iPSCs) and neurite growth in iPSC-derived neurons, whereas PSAP exclusively stimulated neurite growth. CONCLUSION The non-canonical neurotrophic factors MDK, PTN, and PSAP were upregulated in the subacute phase of TBI and played a crucial role in neuroregeneration.
Collapse
Affiliation(s)
- Xuecheng Qiu
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular BiologyXuzhou Medical UniversityXuzhouJiangsuChina
| | - Yaling Guo
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular BiologyXuzhou Medical UniversityXuzhouJiangsuChina
| | - Ming‐Feng Liu
- Department of NeurosurgeryXuzhou Hospital of Traditional Chinese MedicineXuzhouJiangsuChina
| | - Bingge Zhang
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular BiologyXuzhou Medical UniversityXuzhouJiangsuChina
| | - Jingzhen Li
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular BiologyXuzhou Medical UniversityXuzhouJiangsuChina
| | - Jian‐Feng Wei
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular BiologyXuzhou Medical UniversityXuzhouJiangsuChina
- Department of Histology and EmbryologyXuzhou Medical UniversityXuzhouJiangsuChina
| | - Meng Li
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular BiologyXuzhou Medical UniversityXuzhouJiangsuChina
| |
Collapse
|
17
|
Li Z, Liu Y, Huang Y, Tan Q, Mei H, Zhu G, Liu K, Yang G. Circ_0000888 regulates osteogenic differentiation of periosteal mesenchymal stem cells in congenital pseudarthrosis of the tibia. iScience 2023; 26:107923. [PMID: 37810257 PMCID: PMC10551655 DOI: 10.1016/j.isci.2023.107923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 06/02/2023] [Accepted: 09/12/2023] [Indexed: 10/10/2023] Open
Abstract
Congenital pseudarthrosis of the tibia (CPT) is a refractory condition characterized by the decreased osteogenic ability in tibial pseudarthrosis repair. Periosteal mesenchymal stem cells (PMSCs) are multipotent cells involved in bone formation regulation. However, the mechanisms underlying its role in CPT remain unclear. In this study, we observed downregulation of circ_0000888 and pleiotrophin (PTN), as well as upregulation of miR-338-3p in CPT derived PMSCs (CPT-dPMSCs). Our results demonstrated that circ_0000888 and PTN likely enhanced the viability, proliferation, and osteogenic ability of PMSCs, while miR-338-3p had the opposite effect. Further analysis confirmed the regulatory relationship circ_0000888 suppressed the activity of miR-338-3p and upregulated the expression of its downstream target PTN by binding to miR-338-3p, consequently promoting the viability and osteogenic differentiation ability of CPT-dPMSCs. Our findings unveil an unexpected link between circ_0000888/miR-338-3p/PTN in promoting osteogenic ability and indicate the potential pathogenic mechanisms of CPT.
Collapse
Affiliation(s)
- Zhuoyang Li
- Department of Orthopedics, Hunan Provincial Key Laboratory of Pediatric Orthopedics, Hunan Children’s Hospital, Changsha, Hunan, China
- Department of Orthopedics, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yaoxi Liu
- Department of Orthopedics, Hunan Provincial Key Laboratory of Pediatric Orthopedics, Hunan Children’s Hospital, Changsha, Hunan, China
| | - Yiyong Huang
- Department of Orthopedics, Hunan Provincial Key Laboratory of Pediatric Orthopedics, Hunan Children’s Hospital, Changsha, Hunan, China
| | - Qian Tan
- Department of Orthopedics, Hunan Provincial Key Laboratory of Pediatric Orthopedics, Hunan Children’s Hospital, Changsha, Hunan, China
| | - Haibo Mei
- Department of Orthopedics, Hunan Provincial Key Laboratory of Pediatric Orthopedics, Hunan Children’s Hospital, Changsha, Hunan, China
| | - Guanghui Zhu
- Department of Orthopedics, Hunan Provincial Key Laboratory of Pediatric Orthopedics, Hunan Children’s Hospital, Changsha, Hunan, China
| | - Kun Liu
- Department of Orthopedics, Hunan Provincial Key Laboratory of Pediatric Orthopedics, Hunan Children’s Hospital, Changsha, Hunan, China
| | - Ge Yang
- Department of Orthopedics, Hunan Provincial Key Laboratory of Pediatric Orthopedics, Hunan Children’s Hospital, Changsha, Hunan, China
| |
Collapse
|
18
|
Said EA, Al-Dughaishi S, Al-Hatmi W, Al-Reesi I, Al-Balushi MS, Al-Bimani A, Al-Busaidi JZ, Al-Riyami M, Al-Khabori M, Al-Kindi S, Procopio FA, Al-Sinawi S, Al-Ansari A, Koh CY, Al-Naamani K, Al-Jabri AA. Differential Production of Midkine and Pleiotrophin by Innate APCs upon Stimulation through Nucleic Acid-Sensing TLRs. J Immunol Res 2023; 2023:7944102. [PMID: 37850119 PMCID: PMC10578979 DOI: 10.1155/2023/7944102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 07/26/2023] [Accepted: 08/26/2023] [Indexed: 10/19/2023] Open
Abstract
Midkine (MK) and pleiotrophin (PTN) belong to the same family of cytokines. They have similar sequences and functions. Both have important roles in cellular proliferation, tumors, and diseases. They regulate and are expressed by some immune cells. We have recently demonstrated MK production by some human innate antigen-presenting cells (iAPCs), i.e., monocyte-derived dendritic cells (MDDCs) and macrophages stimulated through Toll-like receptor (TLR)-4, and plasmacytoid dendritic cells (pDCs) stimulated through TLR 7. While PTN production was only documented in tissue macrophages. TLRs 3, 7, 8, and 9 are nucleic acid sensing (NAS) TLRs that detect nucleic acids from cell damage and infection and induce iAPC responses. We investigated whether NAS TLRs can induce MK and PTN production by human iAPCs, namely monocytes, macrophages, MDDCs, myeloid dendritic cells (mDCs), and pDCs. Our results demonstrated for the first time that PTN is produced by all iAPCs upon TLR triggering (p < 0.01). IAPCs produced more PTN than MK (p < 0.01). NAS TLRs and iAPCs had differential abilities to induce the production of MK, which was induced in monocytes and pDCs by all NAS TLRs (p < 0.05) and in MDDCs by TLRs 7/8 (p < 0.05). TLR4 induced a stronger MK production than NAS TLRs (p ≤ 0.05). Monocytes produced higher levels of PTN after differentiation to macrophages and MDDCs (p < 0.05). The production of MK and PTN differs among iAPCs, with a higher production of PTN and a selective induction of MK production by NAS TLR. This highlights the potentially important role of iAPCs in angiogenesis, tumors, infections, and autoimmunity through the differential production of MK and PTN upon TLR triggering.
Collapse
Affiliation(s)
- Elias A. Said
- Department of Microbiology and Immunology, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Sumaya Al-Dughaishi
- Department of Microbiology and Immunology, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Wadha Al-Hatmi
- Department of Microbiology and Immunology, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Iman Al-Reesi
- Department of Microbiology and Immunology, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Mohammed S. Al-Balushi
- Department of Microbiology and Immunology, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Atika Al-Bimani
- Department of Microbiology and Immunology, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Juma Z. Al-Busaidi
- Department of Microbiology and Immunology, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Marwa Al-Riyami
- Department of Pathology, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Murtadha Al-Khabori
- Department of Hematology, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Salam Al-Kindi
- Department of Hematology, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Francesco A. Procopio
- Service of Immunology and Allergy, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Shadia Al-Sinawi
- Department of Pathology, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Aliyaa Al-Ansari
- Department of Biology, College of Science, Sultan Qaboos University, Muscat, Oman
| | - Crystal Y. Koh
- Department of Microbiology and Immunology, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | | | - Ali A. Al-Jabri
- Department of Microbiology and Immunology, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| |
Collapse
|
19
|
Abdel-Hafiz HA, Kailasam Mani SK, Huang W, Gouin KH, Chang Y, Xiao T, Ma Q, Li Z, Knott SR, Theodorescu D. Single-cell profiling of murine bladder cancer identifies sex-specific transcriptional signatures with prognostic relevance. iScience 2023; 26:107703. [PMID: 37701814 PMCID: PMC10494466 DOI: 10.1016/j.isci.2023.107703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/18/2023] [Accepted: 08/21/2023] [Indexed: 09/14/2023] Open
Abstract
Bladder cancer (BLCA) is more common in men but more aggressive in women. Sex-based differences in cancer biology are commonly studied using a murine model with BLCA generated by N-butyl-N-(4-hydroxybutyl)-nitrosamine (BBN). While tumors in the BBN model have been profiled, these profiles provide limited information on the tumor microenvironment. Here, we applied single-cell RNA sequencing to characterize cell-type specific transcriptional differences between male and female BBN-induced tumors. We found proportional and gene expression differences in epithelial and non-epithelial subpopulations between male and female tumors. Expression of several genes predicted sex-specific survival in several human BLCA datasets. We identified novel and clinically relevant sex-specific transcriptional signatures including immune cells in the tumor microenvironment and it validated the relevance of the BBN model for studying sex differences in human BLCA. This work highlights the importance of considering sex as a biological variable in the development of new and accurate cancer markers.
Collapse
Affiliation(s)
- Hany A. Abdel-Hafiz
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Cedars-Sinai Samuel Oschin Comprehensive Cancer Institute, Los Angeles, CA, USA
| | | | - Wesley Huang
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Kenneth H. Gouin
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Yuzhou Chang
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center – The James, Columbus, OH 43210, USA
| | - Tong Xiao
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center – The James, Columbus, OH 43210, USA
| | - Qin Ma
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center – The James, Columbus, OH 43210, USA
| | - Zihai Li
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center – The James, Columbus, OH 43210, USA
| | - Simon R.V. Knott
- Cedars-Sinai Samuel Oschin Comprehensive Cancer Institute, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Dan Theodorescu
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Cedars-Sinai Samuel Oschin Comprehensive Cancer Institute, Los Angeles, CA, USA
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
20
|
Farrugia BL, Melrose J. The Glycosaminoglycan Side Chains and Modular Core Proteins of Heparan Sulphate Proteoglycans and the Varied Ways They Provide Tissue Protection by Regulating Physiological Processes and Cellular Behaviour. Int J Mol Sci 2023; 24:14101. [PMID: 37762403 PMCID: PMC10531531 DOI: 10.3390/ijms241814101] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/03/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
This review examines the roles of HS-proteoglycans (HS-PGs) in general, and, in particular, perlecan and syndecan as representative examples and their interactive ligands, which regulate physiological processes and cellular behavior in health and disease. HS-PGs are essential for the functional properties of tissues both in development and in the extracellular matrix (ECM) remodeling that occurs in response to trauma or disease. HS-PGs interact with a biodiverse range of chemokines, chemokine receptors, protease inhibitors, and growth factors in immune regulation, inflammation, ECM stabilization, and tissue protection. Some cell regulatory proteoglycan receptors are dually modified hybrid HS/CS proteoglycans (betaglycan, CD47). Neurexins provide synaptic stabilization, plasticity, and specificity of interaction, promoting neurotransduction, neurogenesis, and differentiation. Ternary complexes of glypican-1 and Robbo-Slit neuroregulatory proteins direct axonogenesis and neural network formation. Specific neurexin-neuroligin complexes stabilize synaptic interactions and neural activity. Disruption in these interactions leads to neurological deficits in disorders of functional cognitive decline. Interactions with HS-PGs also promote or inhibit tumor development. Thus, HS-PGs have complex and diverse regulatory roles in the physiological processes that regulate cellular behavior and the functional properties of normal and pathological tissues. Specialized HS-PGs, such as the neurexins, pikachurin, and Eyes-shut, provide synaptic stabilization and specificity of neural transduction and also stabilize the axenome primary cilium of phototoreceptors and ribbon synapse interactions with bipolar neurons of retinal neural networks, which are essential in ocular vision. Pikachurin and Eyes-Shut interactions with an α-dystroglycan stabilize the photoreceptor synapse. Novel regulatory roles for HS-PGs controlling cell behavior and tissue function are expected to continue to be uncovered in this fascinating class of proteoglycan.
Collapse
Affiliation(s)
- Brooke L. Farrugia
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Melbourne, Melbourne, VIC 3010, Australia;
| | - James Melrose
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
- Raymond Purves Laboratory of Bone and Joint Research, Kolling Institute of Medical Research, Northern Sydney Local Health District, Royal North Shore Hospital, St. Leonards, NSW 2065, Australia
- Sydney Medical School (Northern), University of Sydney at Royal North Shore Hospital, St. Leonards, NSW 2065, Australia
| |
Collapse
|
21
|
Ganguly D, Schmidt MO, Coleman M, Ngo TVC, Sorrelle N, Dominguez AT, Murimwa GZ, Toombs JE, Lewis C, Fang YV, Valdes-Mora F, Gallego-Ortega D, Wellstein A, Brekken RA. Pleiotrophin drives a prometastatic immune niche in breast cancer. J Exp Med 2023; 220:e20220610. [PMID: 36828390 PMCID: PMC9998964 DOI: 10.1084/jem.20220610] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 11/21/2022] [Accepted: 01/09/2023] [Indexed: 11/04/2022] Open
Abstract
Metastatic cancer cells adapt to thrive in secondary organs. To investigate metastatic adaptation, we performed transcriptomic analysis of metastatic and non-metastatic murine breast cancer cells. We found that pleiotrophin (PTN), a neurotrophic cytokine, is a metastasis-associated factor that is expressed highly by aggressive breast cancers. Moreover, elevated PTN in plasma correlated significantly with metastasis and reduced survival of breast cancer patients. Mechanistically, we find that PTN activates NF-κB in cancer cells leading to altered cytokine production, subsequent neutrophil recruitment, and an immune suppressive microenvironment. Consequently, inhibition of PTN, pharmacologically or genetically, reduces the accumulation of tumor-associated neutrophils and reverts local immune suppression, resulting in increased T cell activation and attenuated metastasis. Furthermore, inhibition of PTN significantly enhanced the efficacy of immune checkpoint blockade and chemotherapy in reducing metastatic burden in mice. These findings establish PTN as a previously unrecognized driver of a prometastatic immune niche and thus represents a promising therapeutic target for the treatment of metastatic breast cancer.
Collapse
Affiliation(s)
- Debolina Ganguly
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Cancer Biology Graduate Program, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Marcel O. Schmidt
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Morgan Coleman
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Tuong-Vi Cindy Ngo
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Noah Sorrelle
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Adrian T.A. Dominguez
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Gilbert Z. Murimwa
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jason E. Toombs
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Cheryl Lewis
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yisheng V. Fang
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Fatima Valdes-Mora
- Cancer Epigenetic Biology and Therapeutics group, Precision Medicine Theme, Children’s Cancer Institute, Sydney, Australia
- School of Clinical Medicine, University of NSW Sydney, Sydney, Australia
| | - David Gallego-Ortega
- School of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Technology Sydney, Sydney, Australia
- Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Sydney, Australia
- School of Clinical Medicine, St Vincent’s Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW Sydney, Sydney, Australia
| | - Anton Wellstein
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Rolf A. Brekken
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Cancer Biology Graduate Program, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
22
|
Räuber S, Schroeter CB, Strippel C, Nelke C, Ruland T, Dik A, Golombeck KS, Regner-Nelke L, Paunovic M, Esser D, Münch C, Rosenow F, van Duijn M, Henes A, Ruck T, Amit I, Leypoldt F, Titulaer MJ, Wiendl H, Meuth SG, Meyer Zu Hörste G, Melzer N. Cerebrospinal fluid proteomics indicates immune dysregulation and neuronal dysfunction in antibody associated autoimmune encephalitis. J Autoimmun 2023; 135:102985. [PMID: 36621173 DOI: 10.1016/j.jaut.2022.102985] [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: 10/15/2022] [Accepted: 12/15/2022] [Indexed: 01/09/2023]
Abstract
Autoimmune Encephalitis (AE) spans a group of non-infectious inflammatory conditions of the central nervous system due to an imbalanced immune response. Aiming to elucidate the pathophysiological mechanisms of AE, we applied an unsupervised proteomic approach to analyze the cerebrospinal fluid (CSF) protein profile of AE patients with autoantibodies against N-methyl-d-aspartate receptor (NMDAR) (n = 9), leucine-rich glioma-inactivated protein 1 (LGI1) (n = 9), or glutamate decarboxylase 65 (GAD65) (n = 8) compared to 9 patients with relapsing-remitting multiple sclerosis as inflammatory controls, and 10 patients with somatic symptom disorder as non-inflammatory controls. We found a dysregulation of the complement system, a disbalance between pro-inflammatory and anti-inflammatory proteins on the one hand, and dysregulation of proteins involved in synaptic transmission, synaptogenesis, brain connectivity, and neurodegeneration on the other hand to a different extent in all AE subtypes compared to non-inflammatory controls. Furthermore, elevated levels of several proteases and reduction in protease inhibitors could be detected in all AE subtypes compared to non-inflammatory controls. Moreover, the different AE subtypes showed distinct protein profiles compared to each other and inflammatory controls which may facilitate future identification of disease-specific biomarkers. Overall, CSF proteomics provides insights into the complex pathophysiological mechanisms of AE, including immune dysregulation, neuronal dysfunction, neurodegeneration, and altered protease function.
Collapse
Affiliation(s)
- Saskia Räuber
- Department of Neurology with Institute of Translational Neurology, University of Münster, 48149, Münster, Germany; Department of Neurology, Medical Faculty, Heinrich Heine University of Düsseldorf, 40225 Düsseldorf, Germany
| | - Christina B Schroeter
- Department of Neurology with Institute of Translational Neurology, University of Münster, 48149, Münster, Germany; Department of Neurology, Medical Faculty, Heinrich Heine University of Düsseldorf, 40225 Düsseldorf, Germany
| | - Christine Strippel
- Department of Neurology with Institute of Translational Neurology, University of Münster, 48149, Münster, Germany
| | - Christopher Nelke
- Department of Neurology with Institute of Translational Neurology, University of Münster, 48149, Münster, Germany; Department of Neurology, Medical Faculty, Heinrich Heine University of Düsseldorf, 40225 Düsseldorf, Germany
| | - Tillmann Ruland
- Department of Psychiatry, University of Münster, 48149, Münster, Germany; Department of Psychiatry, Maria Brunn Hospital, 48163, Münster, Germany
| | - Andre Dik
- Department of Neurology with Institute of Translational Neurology, University of Münster, 48149, Münster, Germany; Department of Neurology, Medical Faculty, Heinrich Heine University of Düsseldorf, 40225 Düsseldorf, Germany
| | - Kristin S Golombeck
- Department of Neurology with Institute of Translational Neurology, University of Münster, 48149, Münster, Germany; Department of Neurology, Medical Faculty, Heinrich Heine University of Düsseldorf, 40225 Düsseldorf, Germany
| | - Liesa Regner-Nelke
- Department of Neurology with Institute of Translational Neurology, University of Münster, 48149, Münster, Germany; Department of Neurology, Medical Faculty, Heinrich Heine University of Düsseldorf, 40225 Düsseldorf, Germany
| | - Manuela Paunovic
- Department of Neurology, Erasmus MC University Medical Center, 3015 GD, Rotterdam, the Netherlands
| | - Daniela Esser
- Institute of Clinical Chemistry, University Hospital Schleswig-Holstein, 24105, Kiel, Lübeck, Germany
| | - Christian Münch
- Institute of Biochemistry II, Goethe University Frankfurt, Faculty of Medicine, Theodor-Stern-Kai 7, Building 75, 60590, Frankfurt am Main, Germany; Frankfurt Cancer Institute, Frankfurt am Main, Germany; Cardio-Pulmonary Institute, Frankfurt am Main, Germany
| | - Felix Rosenow
- Epilepsy Center Frankfurt Rhine-Main, Center of Neurology and Neurosurgery, University Hospital Frankfurt, Goethe University Frankfurt, 60528 Frankfurt am Main, Germany; LOEWE Center for Personalized Translational Epilepsy Research (CePTER), Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Martijn van Duijn
- Department of Neurology, Erasmus MC University Medical Center, 3015 GD, Rotterdam, the Netherlands
| | - Antonia Henes
- Department of Neurology, Medical Faculty, Heinrich Heine University of Düsseldorf, 40225 Düsseldorf, Germany
| | - Tobias Ruck
- Department of Neurology with Institute of Translational Neurology, University of Münster, 48149, Münster, Germany; Department of Neurology, Medical Faculty, Heinrich Heine University of Düsseldorf, 40225 Düsseldorf, Germany
| | - Ido Amit
- Department of Immunology, Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Frank Leypoldt
- Institute of Clinical Chemistry, University Hospital Schleswig-Holstein, 24105, Kiel, Lübeck, Germany; Department of Neurology, Faculty of Medicine, Kiel University, 24105, Kiel, Germany
| | - Maarten J Titulaer
- Department of Neurology, Erasmus MC University Medical Center, 3015 GD, Rotterdam, the Netherlands
| | - Heinz Wiendl
- Department of Neurology with Institute of Translational Neurology, University of Münster, 48149, Münster, Germany
| | - Sven G Meuth
- Department of Neurology with Institute of Translational Neurology, University of Münster, 48149, Münster, Germany; Department of Neurology, Medical Faculty, Heinrich Heine University of Düsseldorf, 40225 Düsseldorf, Germany
| | - Gerd Meyer Zu Hörste
- Department of Neurology with Institute of Translational Neurology, University of Münster, 48149, Münster, Germany
| | - Nico Melzer
- Department of Neurology with Institute of Translational Neurology, University of Münster, 48149, Münster, Germany; Department of Neurology, Medical Faculty, Heinrich Heine University of Düsseldorf, 40225 Düsseldorf, Germany.
| |
Collapse
|
23
|
Huang L, Zhang J, Zhang P, Huang X, Yang W, Liu R, Sun Q, Lu Y, Zhang M, Fu Q. Single-cell RNA sequencing uncovers dynamic roadmap and cell-cell communication during buffalo spermatogenesis. iScience 2022; 26:105733. [PMID: 36582818 PMCID: PMC9793287 DOI: 10.1016/j.isci.2022.105733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 10/24/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
Spermatogenesis carries the task of precise intergenerational transmission of genetic information from the paternal genome and involves complex developmental processes regulated by the testicular microenvironment. Studies performed mainly in mouse models have established the theoretical basis for spermatogenesis, yet the wide interspecies differences preclude direct translation of the findings, and farm animal studies are progressing slowly. More than 32,000 cells from prepubertal (3-month-old) and pubertal (24-month-old) buffalo testes were analyzed by using single-cell RNA sequencing (scRNA-seq), and dynamic gene expression roadmaps of germ and somatic cell development were generated. In addition to identifying the dynamic processes of sequential cell fate transitions, the global cell-cell communication essential to maintain regular spermatogenesis in the buffalo testicular microenvironment was uncovered. The findings provide the theoretical basis for establishing buffalo germline stem cells in vitro or culturing organoids and facilitating the expansion of superior livestock breeding.
Collapse
Affiliation(s)
- Liangfeng Huang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Junjun Zhang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Pengfei Zhang
- Institute of Medical and Health, Guangxi Academy of Sciences, Nanning 530007, China
| | - Xingchen Huang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Weihan Yang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Runfeng Liu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Qinqiang Sun
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Yangqing Lu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, China,Corresponding author
| | - Ming Zhang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, China,Corresponding author
| | - Qiang Fu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science and Technology, Guangxi University, Nanning 530004, China,Corresponding author
| |
Collapse
|
24
|
Shu S, Fu M, Chen X, Zhang N, Zhao R, Chang Y, Cui H, Liu Z, Wang X, Hua X, Li Y, Wang X, Wang X, Feng W, Song J. Cellular Landscapes of Nondiseased Human Cardiac Valves From End-Stage Heart Failure-Explanted Heart. Arterioscler Thromb Vasc Biol 2022; 42:1429-1446. [PMID: 36200446 DOI: 10.1161/atvbaha.122.318314] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Exploring the mechanisms of valvular heart disease at the cellular level may be useful to identify new therapeutic targets; however, the comprehensive cellular landscape of nondiseased human cardiac valve leaflets remains unclear. METHODS The cellular landscapes of nondiseased human cardiac valve leaflets (5 aortic valves, 5 pulmonary valves, 5 tricuspid valves, and 3 mitral valves) from end-stage heart failure patients undergoing heart transplantation were explored using single-cell RNA sequencing. Bioinformatics was used to identify the cell types, describe the cell functions, and investigate cellular developmental trajectories and interactions. Differences among the 4 types of cardiac valves at the cellular level were summarized. Pathological staining was performed to validate the key findings of single-cell RNA sequencing. An integrative analysis of our single-cell data and published genome-wide association study-based and bulk RNA sequencing-based data provided insights into the cell-specific contributions to calcific aortic valve diseases. RESULTS Six cell types were identified among 128 412 cells from nondiseased human cardiac valve leaflets. Valvular interstitial cells were the largest population, followed by myeloid cells, lymphocytes, valvular endothelial cells, mast cells, and myofibroblasts. The 4 types of cardiac valve had distinct cellular compositions. The intercellular communication analysis revealed that valvular interstitial cells were at the center of the communication network. The integrative analysis of our single-cell RNA sequencing data revealed key cellular subpopulations involved in the pathogenesis of calcific aortic valve diseases. CONCLUSIONS The cellular landscape differed among the 4 types of nondiseased cardiac valve, which might explain their differences in susceptibility to pathological remodeling and valvular heart disease.
Collapse
Affiliation(s)
- Songren Shu
- State Key Laboratory of Cardiovascular Disease (S.S., M.F., X.C., N.Z., R.Z., Y.C., H.C., Z.L., Xiaohu Wang, X.H., Xin Wang, Xianqiang Wang, W.F., J.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,The Cardiomyopathy Research Group (S.S., M.F., X.C., Y.C., H.C., Z.L., X.H., J.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mengxia Fu
- State Key Laboratory of Cardiovascular Disease (S.S., M.F., X.C., N.Z., R.Z., Y.C., H.C., Z.L., Xiaohu Wang, X.H., Xin Wang, Xianqiang Wang, W.F., J.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,The Cardiomyopathy Research Group (S.S., M.F., X.C., Y.C., H.C., Z.L., X.H., J.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiao Chen
- State Key Laboratory of Cardiovascular Disease (S.S., M.F., X.C., N.Z., R.Z., Y.C., H.C., Z.L., Xiaohu Wang, X.H., Xin Wang, Xianqiang Wang, W.F., J.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,The Cardiomyopathy Research Group (S.S., M.F., X.C., Y.C., H.C., Z.L., X.H., J.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ningning Zhang
- State Key Laboratory of Cardiovascular Disease (S.S., M.F., X.C., N.Z., R.Z., Y.C., H.C., Z.L., Xiaohu Wang, X.H., Xin Wang, Xianqiang Wang, W.F., J.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ruojin Zhao
- State Key Laboratory of Cardiovascular Disease (S.S., M.F., X.C., N.Z., R.Z., Y.C., H.C., Z.L., Xiaohu Wang, X.H., Xin Wang, Xianqiang Wang, W.F., J.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuan Chang
- State Key Laboratory of Cardiovascular Disease (S.S., M.F., X.C., N.Z., R.Z., Y.C., H.C., Z.L., Xiaohu Wang, X.H., Xin Wang, Xianqiang Wang, W.F., J.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,The Cardiomyopathy Research Group (S.S., M.F., X.C., Y.C., H.C., Z.L., X.H., J.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hao Cui
- State Key Laboratory of Cardiovascular Disease (S.S., M.F., X.C., N.Z., R.Z., Y.C., H.C., Z.L., Xiaohu Wang, X.H., Xin Wang, Xianqiang Wang, W.F., J.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,The Cardiomyopathy Research Group (S.S., M.F., X.C., Y.C., H.C., Z.L., X.H., J.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zirui Liu
- State Key Laboratory of Cardiovascular Disease (S.S., M.F., X.C., N.Z., R.Z., Y.C., H.C., Z.L., Xiaohu Wang, X.H., Xin Wang, Xianqiang Wang, W.F., J.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,The Cardiomyopathy Research Group (S.S., M.F., X.C., Y.C., H.C., Z.L., X.H., J.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaohu Wang
- State Key Laboratory of Cardiovascular Disease (S.S., M.F., X.C., N.Z., R.Z., Y.C., H.C., Z.L., Xiaohu Wang, X.H., Xin Wang, Xianqiang Wang, W.F., J.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiumeng Hua
- State Key Laboratory of Cardiovascular Disease (S.S., M.F., X.C., N.Z., R.Z., Y.C., H.C., Z.L., Xiaohu Wang, X.H., Xin Wang, Xianqiang Wang, W.F., J.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,The Cardiomyopathy Research Group (S.S., M.F., X.C., Y.C., H.C., Z.L., X.H., J.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuan Li
- Department of Cardiovascular Surgery (Y.L., Xin Wang, Xianqiang Wang, W.F., J.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xin Wang
- State Key Laboratory of Cardiovascular Disease (S.S., M.F., X.C., N.Z., R.Z., Y.C., H.C., Z.L., Xiaohu Wang, X.H., Xin Wang, Xianqiang Wang, W.F., J.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Department of Cardiovascular Surgery (Y.L., Xin Wang, Xianqiang Wang, W.F., J.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xianqiang Wang
- State Key Laboratory of Cardiovascular Disease (S.S., M.F., X.C., N.Z., R.Z., Y.C., H.C., Z.L., Xiaohu Wang, X.H., Xin Wang, Xianqiang Wang, W.F., J.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Department of Cardiovascular Surgery (Y.L., Xin Wang, Xianqiang Wang, W.F., J.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Feng
- State Key Laboratory of Cardiovascular Disease (S.S., M.F., X.C., N.Z., R.Z., Y.C., H.C., Z.L., Xiaohu Wang, X.H., Xin Wang, Xianqiang Wang, W.F., J.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Department of Cardiovascular Surgery (Y.L., Xin Wang, Xianqiang Wang, W.F., J.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiangping Song
- State Key Laboratory of Cardiovascular Disease (S.S., M.F., X.C., N.Z., R.Z., Y.C., H.C., Z.L., Xiaohu Wang, X.H., Xin Wang, Xianqiang Wang, W.F., J.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,The Cardiomyopathy Research Group (S.S., M.F., X.C., Y.C., H.C., Z.L., X.H., J.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Department of Cardiovascular Surgery (Y.L., Xin Wang, Xianqiang Wang, W.F., J.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen' China (J.S.)
| |
Collapse
|
25
|
Li R, Wang TY, Shelp-Peck E, Wu SP, DeMayo FJ. The single-cell atlas of cultured human endometrial stromal cells. F&S SCIENCE 2022; 3:349-366. [PMID: 36089208 PMCID: PMC9669198 DOI: 10.1016/j.xfss.2022.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 09/01/2022] [Accepted: 09/02/2022] [Indexed: 06/15/2023]
Abstract
OBJECTIVE To systematically analyze the cell composition and transcriptome of primary human endometrial stromal cells (HESCs) and transformed human endometrial stromal cells (THESCs). DESIGN The primary HESCs from 3 different donors and 1 immortalized THESC were collected from the human endometrium at the midsecretory phase and cultured in vitro. SETTING Academic research laboratory. PATIENT(S) None. INTERVENTION(S) None. MAIN OUTCOME MEASURE(S) Single-cell ribonucleic acid sequencing analysis. RESULT(S) We found the individual differences among the primary HESCs and bigger changes between the primary HESCs and THESCs. Cell clustering with or without integration identified cell clusters belonging to mature, proliferative, and active fibroblasts that were conserved across all samples at different stages of the cell cycles with intensive cell communication signals. All primary HESCs and THESCs can be correlated with some subpopulations of fibroblasts in the human endometrium. CONCLUSION(S) Our study indicated that the primary HESCs and THESCs displayed conserved cell characters and distinct cell clusters. Mature, proliferative, and active fibroblasts at different stages or cell cycles were detected across all samples and presented with a complex cell communication network. The cultured HESCs and THESCs retained the features of some subpopulations within the human endometrium.
Collapse
Affiliation(s)
- Rong Li
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, North Carolina
| | - Tian-Yuan Wang
- Integrative Bioinformatics, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, North Carolina
| | - Elinor Shelp-Peck
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, North Carolina; The Biological Sciences Department, The Department of Chemistry, Physics, and Geosciences, Meredith College, Raleigh, North Carolina
| | - San-Pin Wu
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, North Carolina
| | - Francesco J DeMayo
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, North Carolina.
| |
Collapse
|
26
|
Gong T, Wang Y, Dong S, Ma X, Du D, Zou C, Zheng Q, Wen Z. Single-cell RNA-seq reveals the communications between extracellular matrix-related components and Schwann cells contributing to the earlobe keloid formation. Front Med (Lausanne) 2022; 9:1000324. [PMID: 36388926 PMCID: PMC9643690 DOI: 10.3389/fmed.2022.1000324] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 09/28/2022] [Indexed: 07/26/2023] Open
Abstract
Keloid is a major type of skin fibrotic disease, with one prominent feature of extensive accumulation of extracellular matrix (ECM) components, and another feature of pain/itching, which is closely related to the peripheral nervous system (PNS). However, the molecular pathogenesis of these two prominent features still needs to be further explored. In the present study, we performed single-cell RNA sequencing (scRNA-seq) on clinical earlobe keloid samples and adjacent normal skin samples and constructed a keloid atlas of 31,379 cells. All cells were clustered into 13 major cell types using cell-type-specific markers. Among them, fibroblast, vascular endothelial cells, and smooth muscle cells were defined as the ECM-related populations according to their ECM-associated functions. Also, we found that Schwann cells (SCs) were the main neuron cells of PNS in the skin. Interestingly, the cell proportions of ECM-related populations, as well as SC were increased significantly in the earlobe keloid compared to the adjacent normal tissues, suggesting an important role of these cell types in the development of the earlobe keloid. Comprehensive cell-cell interaction analysis at the single-cell level revealed a strong interaction between SC and ECM-related subgroups which might be mediated by SEMA3C signaling pathways and MK/PTN gene family, which are found to be mainly involved in promoting cell proliferation and migration. Moreover, further exploration of the interactions of ECM-related populations and SC in different keloids, including earlobe keloid, back keloid, and chest keloid revealed an increasing amount of TGFβ-TGFβ receptor interactions in chest/back keloids as compared to earlobe keloid, which suggested the anatomic site-specific pathogenesis in different keloids. Altogether, these findings suggested the interactions between ECM-related populations and SC contributing to the earlobe keloid formation and helped us to better understand the pathogenesis of keloids.
Collapse
Affiliation(s)
- Taogen Gong
- Otolaryngology-Head and Neck Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Department of Otolaryngology-Head and Neck Surgery, The Second Clinical Medical College, Jinan University (Shenzhen People’s Hospital), Shenzhen, China
| | - Yayu Wang
- Department of Cardiovascular Surgery, The Second Clinical Medical College, Jinan University (Shenzhen People’s Hospital), Shenzhen, China
| | - Shaowei Dong
- School of Medicine, Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, China
| | - Xiaoshi Ma
- Department of Pathology, The Second Clinical Medical College, Jinan University (Shenzhen People’s Hospital), Shenzhen, China
| | - Danfeng Du
- Department of Pathology, The Second Clinical Medical College, Jinan University (Shenzhen People’s Hospital), Shenzhen, China
| | - Chang Zou
- School of Medicine, Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, China
| | - Qijun Zheng
- Department of Cardiovascular Surgery, The Second Clinical Medical College, Jinan University (Shenzhen People’s Hospital), Shenzhen, China
| | - Zhong Wen
- Otolaryngology-Head and Neck Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
27
|
Pan-Cancer Analysis of the Prognostic and Immunotherapeutic Value of MITD1. Cells 2022; 11:cells11203308. [PMID: 36291174 PMCID: PMC9600621 DOI: 10.3390/cells11203308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 10/12/2022] [Accepted: 10/19/2022] [Indexed: 11/17/2022] Open
Abstract
Microtubule-interacting and trafficking domain containing 1 (MITD1) is associated with abscission during cytokinesis. However, systematic investigation into its role in cancer is lacking. Therefore, we explored the pan-cancer role of MITD1 using multiple databases. Expression and clinical survival, immunological, and enrichment analyses were performed using R packages and online tools. For breast cancer, single-cell level analysis, immunochemistry, and in vitro experiments were performed to explore the mechanism of MITD1. A nomogram was established to predict the prognosis of patients with breast cancer and evaluate the immunotherapy biomarker based on two datasets. In some cancers, high MITD1 expression was associated with a more favorable prognosis. For instance, it inhibited tumor cell proliferation and migration in breast cancer. MITD1 may regulate cancer development by altering the tumor microenvironment, and MITD1 expression may predict the response to immune checkpoint blockade, platinum, and poly ADP-ribose polymerase inhibitor therapies. Our nomogram was used to determine the prognosis of patients with breast cancer. MITD1 can also predict the response to immunotherapy. Our first pan-cancer study of MITD1 has shown that it plays different roles in cancer development and therapy. In breast cancer, MITD1 inhibited cell proliferation and migration and serves as a new biomarker.
Collapse
|
28
|
Huang Z, Wu C, Liu X, Lu S, You L, Guo F, Stalin A, Zhang J, Zhang F, Wu Z, Tan Y, Fan X, Huang J, Zhai Y, Shi R, Chen M, Wu C, Li H, Wu J. Single-Cell and Bulk RNA Sequencing Reveal Malignant Epithelial Cell Heterogeneity and Prognosis Signatures in Gastric Carcinoma. Cells 2022; 11:cells11162550. [PMID: 36010627 PMCID: PMC9407012 DOI: 10.3390/cells11162550] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/10/2022] [Accepted: 08/12/2022] [Indexed: 11/24/2022] Open
Abstract
Gastric carcinoma (GC) heterogeneity represents a major barrier to accurate diagnosis and treatment. Here, we established a comprehensive single-cell transcriptional atlas to identify the cellular heterogeneity in malignant epithelial cells of GC using single-cell RNA sequencing (scRNA-seq). A total of 49,994 cells from nine patients with paired primary tumor and normal tissues were analyzed by multiple strategies. This study focused on the malignant epithelial cells, which were divided into three subtypes, including pit mucous cells, chief cells, and gastric and intestinal cells. The trajectory analysis results suggest that the differentiation of the three subtypes could be from the pit mucous cells to the chief cells and then to the gastric and intestinal cells. Lauren’s histopathology of GC might originate from various subtypes of malignant epithelial cells. The functional enrichment analysis results show that the three subtypes focused on different biological processes (BP) and pathways related to tumor development. In addition, we generated and validated the prognostic signatures for predicting the OS in GC patients by combining the scRNA-seq and bulk RNA sequencing (bulk RNA-seq) datasets. Overall, our study provides a resource for understanding the heterogeneity of GC that will contribute to accurate diagnosis and prognosis.
Collapse
Affiliation(s)
- Zhihong Huang
- Department of Clinical Pharmacology of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Chao Wu
- Department of Clinical Pharmacology of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xinkui Liu
- Department of Clinical Pharmacology of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Shan Lu
- Department of Clinical Pharmacology of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Leiming You
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Fengying Guo
- School of Management, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Antony Stalin
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Jingyuan Zhang
- Department of Clinical Pharmacology of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Fanqin Zhang
- Department of Clinical Pharmacology of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Zhishan Wu
- Department of Clinical Pharmacology of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yingying Tan
- Department of Clinical Pharmacology of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xiaotian Fan
- Department of Clinical Pharmacology of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jiaqi Huang
- Department of Clinical Pharmacology of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yiyan Zhai
- Department of Clinical Pharmacology of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Rui Shi
- Department of Clinical Pharmacology of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Meilin Chen
- Department of Clinical Pharmacology of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Chunfang Wu
- Department of Operations, Beijing Zest Bridge Medical Technology Inc., Beijing 100176, China
- Correspondence: (C.W.); (J.W.)
| | - Huiying Li
- School of Biology, Beijing Forestry University, Beijing 100091, China
| | - Jiarui Wu
- Department of Clinical Pharmacology of Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
- Correspondence: (C.W.); (J.W.)
| |
Collapse
|
29
|
Li Z, Shen F, Song L, Zhang S. Antifungal Activity of NP20 Derived from Amphioxus Midkine/Pleiotrophin Homolog Against Aspergillus niger and Aspergillus fumigatus. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2022; 24:614-625. [PMID: 35610324 DOI: 10.1007/s10126-022-10131-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 04/26/2022] [Indexed: 06/15/2023]
Abstract
With the emergence of antifungal resistance, systematic infections with Aspergillus are becoming the major cause of the clinical morbidity. The development of novel antifungal agents with high efficacy, low drug tolerance, and few side effects is urgent. In response to that need, we have identified NP20. Here we demonstrate clearly that NP20 has antifungal activity, capable of killing the spores of Aspergillus niger and Aspergillus fumigatus as well as causing direct damage to the surface, membrane, cytoplasm, organelle, and nucleus of the fungal spores. Interestingly, NP20 is active under temperature stress and a wide range of pH. Subsequently, MTT assay, assay for binding of NP20 to fungal cell wall components, membrane depolarization assay, confocal microscopy, ROS assay, DNA replication, and protein synthesis assay are performed to clarify the mechanisms underlying NP20 against Aspergillus. The results show that NP20 can bind with and pass through the fungal cell wall, and then interfere with the lipid membrane. Moreover, NP20 can induce intracellular ROS production, DNA fragmentation, and protein synthesis inhibition of the fungal cells. These together indicate that NP20 is a novel antifungal peptide, which has considerable potential for future development as novel peptide antibiotics against Aspergillus.
Collapse
Affiliation(s)
- Zhi Li
- Laboratory for Evolution & Development, Institute of Evolution & Marine Biodiversity and Department of Marine Biology, Ocean University of China, Room 320, 5 Yushan Road, Darwin Building, Qingdao, 266003, China
| | - Fangwang Shen
- Laboratory for Evolution & Development, Institute of Evolution & Marine Biodiversity and Department of Marine Biology, Ocean University of China, Room 320, 5 Yushan Road, Darwin Building, Qingdao, 266003, China
| | - Lili Song
- Laboratory for Evolution & Development, Institute of Evolution & Marine Biodiversity and Department of Marine Biology, Ocean University of China, Room 320, 5 Yushan Road, Darwin Building, Qingdao, 266003, China
| | - Shicui Zhang
- Laboratory for Evolution & Development, Institute of Evolution & Marine Biodiversity and Department of Marine Biology, Ocean University of China, Room 320, 5 Yushan Road, Darwin Building, Qingdao, 266003, China.
- Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, 266003, China.
| |
Collapse
|
30
|
Xia T, Chen D, Liu X, Qi H, Wang W, Chen H, Ling T, Otkur W, Zhang CS, Kim J, Lin SC, Piao HL. Midkine noncanonically suppresses AMPK activation through disrupting the LKB1-STRAD-Mo25 complex. Cell Death Dis 2022; 13:414. [PMID: 35487917 PMCID: PMC9054788 DOI: 10.1038/s41419-022-04801-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 03/23/2022] [Accepted: 03/30/2022] [Indexed: 11/09/2022]
Abstract
Midkine (MDK), a secreted growth factor, regulates signal transduction and cancer progression by interacting with receptors, and it can be internalized into the cytoplasm by endocytosis. However, its intracellular function and signaling regulation remain unclear. Here, we show that intracellular MDK interacts with LKB1 and STRAD to disrupt the LKB1-STRAD-Mo25 complex. Consequently, MDK decreases the activity of LKB1 to dampen both the basal and stress-induced activation of AMPK by glucose starvation or treatment of 2-DG. We also found that MDK accelerates cancer cell proliferation by inhibiting the activation of the LKB1-AMPK axis. In human cancers, compared to other well-known growth factors, MDK expression is most significantly upregulated in cancers, especially in liver, kidney and breast cancers, correlating with clinical outcomes and inversely correlating with phosphorylated AMPK levels. Our study elucidates an inhibitory mechanism for AMPK activation, which is mediated by the intracellular MDK through disrupting the LKB1-STRAD-Mo25 complex.
Collapse
|
31
|
Li Y, Dittmann NL, Watson AES, de Almeida MMA, Footz T, Voronova A. Hepatoma Derived Growth Factor Enhances Oligodendrocyte Genesis from Subventricular Zone Precursor Cells. ASN Neuro 2022; 14:17590914221086340. [PMID: 35293825 PMCID: PMC8943302 DOI: 10.1177/17590914221086340] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Oligodendrocytes, the myelinating cells of the central nervous system (CNS), perform vital functions in neural protection and communication, as well as cognition. Enhanced production of oligodendrocytes has been identified as a therapeutic approach for neurodegenerative and neurodevelopmental disorders. In the postnatal brain, oligodendrocytes are generated from the neural stem and precursor cells (NPCs) in the subventricular zone (SVZ) and parenchymal oligodendrocyte precursor cells (OPCs). Here, we demonstrate exogenous Hepatoma Derived Growth Factor (HDGF) enhances oligodendrocyte genesis from murine postnatal SVZ NPCs in vitro without affecting neurogenesis or astrogliogenesis. We further show that this is achieved by increasing proliferation of both NPCs and OPCs, as well as OPC differentiation into oligodendrocytes. In vivo results demonstrate that intracerebroventricular infusion of HDGF leads to increased oligodendrocyte genesis from SVZ NPCs, as well as OPC proliferation. Our results demonstrate a novel role for HDGF in regulating SVZ precursor cell proliferation and oligodendrocyte differentiation.
Collapse
Affiliation(s)
- Yutong Li
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, T6G 2H7, Canada
| | - Nicole Leanne Dittmann
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, T6G 2H7, Canada
- Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, T6G 2E1, Canada
| | - Adrianne Eve Scovil Watson
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, T6G 2H7, Canada
| | - Monique Marylin Alves de Almeida
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, T6G 2H7, Canada
- Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, T6G 2E1, Canada
| | - Tim Footz
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, T6G 2H7, Canada
| | - Anastassia Voronova
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, T6G 2H7, Canada
- Neuroscience and Mental Health Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, T6G 2E1, Canada
- Women and Children’s Health Research Institute, 5-083 Edmonton Clinic Health Academy, University of Alberta, 11405 87 Avenue NW Edmonton, Alberta, Canada, T6G 1C9
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, T6G 2H7, Canada
- Multiple Sclerosis Centre, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, T6G 2H7, Canada
| |
Collapse
|
32
|
Casari I, Emmanouilidi A, Domenichini A, Falasca M. Extracellular vesicles derived from pancreatic cancer cells are enriched in the growth factor Midkine. Adv Biol Regul 2021; 83:100857. [PMID: 34916167 DOI: 10.1016/j.jbior.2021.100857] [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: 11/05/2021] [Revised: 12/02/2021] [Accepted: 12/06/2021] [Indexed: 11/26/2022]
Abstract
The growth factor Midkine is a heparin-binding cytokine originally discovered during the differentiation process induced by the retinoic acid in embryonal carcinoma cells. Several studies pointed out the key role of this protein in tumour progression and its elevated expression in different malignancies, including pancreatic cancer. New diagnostic and therapeutic tools are urgently required to treat this highly aggressive and incurable disease capable of metastasising, evading diagnosis, and resisting therapy. Serum midkine promises to be a very functional tumour marker and a target for cancer treatment as an elevated concentration of serum midkine is consistently reported in patients with various tumours. Here, we identified high levels of midkine in extracellular vesicles isolated from pancreatic cancer cell lines and showed that it stimulates the growth of pancreatic cancer cells not expressing midkine.
Collapse
Affiliation(s)
- Ilaria Casari
- Metabolic Signalling Group, Curtin Medical School, Curtin Health Innovation Research Institute, Curtin University, 6102, Perth, WA, Australia
| | - Aikaterini Emmanouilidi
- Metabolic Signalling Group, Curtin Medical School, Curtin Health Innovation Research Institute, Curtin University, 6102, Perth, WA, Australia
| | - Alice Domenichini
- Metabolic Signalling Group, Curtin Medical School, Curtin Health Innovation Research Institute, Curtin University, 6102, Perth, WA, Australia
| | - Marco Falasca
- Metabolic Signalling Group, Curtin Medical School, Curtin Health Innovation Research Institute, Curtin University, 6102, Perth, WA, Australia.
| |
Collapse
|
33
|
Olmeda D, Cerezo‐Wallis D, Mucientes C, Calvo TG, Cañón E, Alonso‐Curbelo D, Ibarz N, Muñoz J, Rodriguez‐Peralto JL, Ortiz‐Romero P, Ortega S, Soengas MS. Live imaging of neolymphangiogenesis identifies acute antimetastatic roles of dsRNA mimics. EMBO Mol Med 2021; 13:e12924. [PMID: 34762341 PMCID: PMC8649872 DOI: 10.15252/emmm.202012924] [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: 06/11/2020] [Revised: 10/12/2021] [Accepted: 10/15/2021] [Indexed: 11/18/2022] Open
Abstract
Long-range communication between tumor cells and the lymphatic vasculature defines competency for metastasis in different cancer types, particularly in melanoma. Nevertheless, the discovery of selective blockers of lymphovascular niches has been compromised by the paucity of experimental systems for whole-body analyses of tumor progression. Here, we exploit immunocompetent and immunodeficient mouse models for live imaging of Vegfr3-driven neolymphangiogenesis, as a versatile platform for drug screening in vivo. Spatiotemporal analyses of autochthonous melanomas and patient-derived xenografts identified double-stranded RNA mimics (dsRNA nanoplexes) as potent inhibitors of neolymphangiogenesis, metastasis, and post-surgical disease relapse. Mechanistically, dsRNA nanoplexes were found to exert a rapid dual action in tumor cells and in their associated lymphatic vasculature, involving the transcriptional repression of the lymphatic drivers Midkine and Vegfr3, respectively. This suppressive function was mediated by a cell-autonomous type I interferon signaling and was not shared by FDA-approved antimelanoma treatments. These results reveal an alternative strategy for targeting the tumor cell-lymphatic crosstalk and underscore the power of Vegfr3-lymphoreporters for pharmacological testing in otherwise aggressive cancers.
Collapse
Affiliation(s)
- David Olmeda
- Melanoma LaboratoryMolecular Oncology ProgrammeSpanish National Cancer Research Centre (CNIO)MadridSpain
| | - Daniela Cerezo‐Wallis
- Melanoma LaboratoryMolecular Oncology ProgrammeSpanish National Cancer Research Centre (CNIO)MadridSpain
- Present address:
Spanish National Center for Cardiovascular Research (CNIC)MadridSpain
| | - Cynthia Mucientes
- Melanoma LaboratoryMolecular Oncology ProgrammeSpanish National Cancer Research Centre (CNIO)MadridSpain
| | - Tonantzin G Calvo
- Melanoma LaboratoryMolecular Oncology ProgrammeSpanish National Cancer Research Centre (CNIO)MadridSpain
| | - Estela Cañón
- Melanoma LaboratoryMolecular Oncology ProgrammeSpanish National Cancer Research Centre (CNIO)MadridSpain
| | - Direna Alonso‐Curbelo
- Melanoma LaboratoryMolecular Oncology ProgrammeSpanish National Cancer Research Centre (CNIO)MadridSpain
- Present address:
Memorial Sloan Kettering Cancer CentreNew YorkNYUSA
| | - Nuria Ibarz
- Proteomics UnitBiotechnology Programme, ProteoRed‐ISCIIISpanish National Cancer Research Centre (CNIO)MadridSpain
| | - Javier Muñoz
- Proteomics UnitBiotechnology Programme, ProteoRed‐ISCIIISpanish National Cancer Research Centre (CNIO)MadridSpain
| | - José L Rodriguez‐Peralto
- Instituto de Investigación i+12Hospital 12 de OctubreUniversidad Complutense Madrid Medical SchoolMadridSpain
| | - Pablo Ortiz‐Romero
- Department of DermatologyHospital 12 de OctubreUniversidad Complutense Madrid Medical SchoolMadridSpain
| | - Sagrario Ortega
- Mouse Genome Editing Core UnitSpanish National Cancer Research Centre (CNIO)MadridSpain
| | - María S Soengas
- Melanoma LaboratoryMolecular Oncology ProgrammeSpanish National Cancer Research Centre (CNIO)MadridSpain
| |
Collapse
|
34
|
Trocha M, Fleszar MG, Fortuna P, Lewandowski Ł, Gostomska-Pampuch K, Sozański T, Merwid-Ląd A, Krzystek-Korpacka M. Sitagliptin Modulates Oxidative, Nitrative and Halogenative Stress and Inflammatory Response in Rat Model of Hepatic Ischemia-Reperfusion. Antioxidants (Basel) 2021; 10:antiox10081168. [PMID: 34439416 PMCID: PMC8388898 DOI: 10.3390/antiox10081168] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 07/19/2021] [Accepted: 07/20/2021] [Indexed: 12/23/2022] Open
Abstract
A possibility of repurposing sitagliptin, a well-established antidiabetic drug, for alleviating injury caused by ischemia-reperfusion (IR) is being researched. The aim of this study was to shed some light on the molecular background of the protective activity of sitagliptin during hepatic IR. The expression and/or concentration of inflammation and oxidative stress-involved factors have been determined in rat liver homogenates using quantitative RT-PCR and Luminex® xMAP® technology and markers of nitrative and halogenative stress were quantified using targeted metabolomics (LC-MS/MS). Animals (n = 36) divided into four groups were treated with sitagliptin (5 mg/kg) (S and SIR) or saline solution (C and IR), and the livers from IR and SIR were subjected to ischemia (60 min) and reperfusion (24 h). The midkine expression (by 2.2-fold) and the free 3-nitrotyrosine (by 2.5-fold) and IL-10 (by 2-fold) concentration were significantly higher and the Nox4 expression was lower (by 9.4-fold) in the IR than the C animals. As compared to IR, the SIR animals had a lower expression of interleukin-6 (by 4.2-fold) and midkine (by 2-fold), a lower concentration of 3-nitrotyrosine (by 2.5-fold) and a higher Nox4 (by 2.9-fold) and 3-bromotyrosine (by 1.4-fold). In conclusion, IR disturbs the oxidative, nitrative and halogenative balance and aggravates the inflammatory response in the liver, which can be attenuated by low doses of sitagliptin.
Collapse
Affiliation(s)
- Małgorzata Trocha
- Department of Pharmacology, Wroclaw Medical University, 50-345 Wroclaw, Poland; (T.S.); (A.M.-L.)
- Correspondence: (M.T.); (M.K.-K.)
| | - Mariusz G. Fleszar
- Department of Biochemistry and Immunochemistry, Wroclaw Medical University, 50-368 Wroclaw, Poland; (M.G.F.); (P.F.); (Ł.L.); (K.G.-P.)
| | - Paulina Fortuna
- Department of Biochemistry and Immunochemistry, Wroclaw Medical University, 50-368 Wroclaw, Poland; (M.G.F.); (P.F.); (Ł.L.); (K.G.-P.)
| | - Łukasz Lewandowski
- Department of Biochemistry and Immunochemistry, Wroclaw Medical University, 50-368 Wroclaw, Poland; (M.G.F.); (P.F.); (Ł.L.); (K.G.-P.)
| | - Kinga Gostomska-Pampuch
- Department of Biochemistry and Immunochemistry, Wroclaw Medical University, 50-368 Wroclaw, Poland; (M.G.F.); (P.F.); (Ł.L.); (K.G.-P.)
| | - Tomasz Sozański
- Department of Pharmacology, Wroclaw Medical University, 50-345 Wroclaw, Poland; (T.S.); (A.M.-L.)
| | - Anna Merwid-Ląd
- Department of Pharmacology, Wroclaw Medical University, 50-345 Wroclaw, Poland; (T.S.); (A.M.-L.)
| | - Małgorzata Krzystek-Korpacka
- Department of Biochemistry and Immunochemistry, Wroclaw Medical University, 50-368 Wroclaw, Poland; (M.G.F.); (P.F.); (Ł.L.); (K.G.-P.)
- Correspondence: (M.T.); (M.K.-K.)
| |
Collapse
|
35
|
Reyes-Mata PM, Rojas-Mayorquín AE, Carrera-Quintanar L, González-Castillo C, Mireles-Ramírez MA, Guerrero-García JDJ, Ortuño-Sahagún D. Pleiotrophin serum level is increased in Relapsing-Remitting Multiple Sclerosis and correlates with sex, BMI and treatment. Arch Med Res 2021; 53:59-68. [PMID: 34247888 DOI: 10.1016/j.arcmed.2021.06.005] [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: 01/11/2021] [Revised: 05/02/2021] [Accepted: 06/15/2021] [Indexed: 11/18/2022]
Abstract
BACKGROUND Multiple Sclerosis (MS) is an immune-mediated demyelinating disease mainly affecting the Central Nervous System (CNS). 80% of MS patients present the Relapsing-Remitting form (RRMS). Pleiotrophin (PTN), a cytokine previously associated with other autoimmune and neurological diseases, could play a role in the pathophysiology of RRMS due to its neuro and immunomodulatory effect. However, PTN has never been explored in RRMS patients. AIM OF THE STUDY To determine PTN serum levels in patients with RRMS, treated with Glatiramer acetate (GA) or Interferon-beta (IFN-β), as well as in non-treated patients and healthy controls as a first attempt to explore PTN in RRMS. METHODS PTN serum levels were quantified by ELISA in 57 patients and 18 controls. RESULTS We demonstrated that PTN serum levels are significantly higher in RRMS patients. In IFN-β treated patients alone, PTN correlated positively with time of disease evolution and time of IFN-β use and correlated negatively with the MS severity score (MSSS). When comparing groups according to weight status, we observed that PTN is statistically increased in overweight female patients and that weight does not affect male patients. The Area Under the Curve (AUC) of the Receiver Operating Characteristic (ROC) curve analysis was higher for males compared to females. CONCLUSION PTN serum level is higher in RRMS patients and that is associated with sex, BMI and IFN-β treatment. Therefore, we propose that PTN could be playing a role in MS. Further studies must be performed to identify the exact role of PTN in this pathology.
Collapse
Affiliation(s)
- Paulina María Reyes-Mata
- Laboratorio de Neuroinmunobiología Molecular, Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Jalisco, México
| | - Argelia Esperanza Rojas-Mayorquín
- Departamento de Ciencias Ambientales, Centro Universitario de Ciencias Biológicas y Agropecuarias, Universidad de Guadalajara, Jalisco, México
| | - Lucrecia Carrera-Quintanar
- Laboratorio de Ciencias de los Alimentos, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Jalisco, México
| | | | - Mario Alberto Mireles-Ramírez
- Unidad Médica de Alta Especialidad, Hospital de Especialidades, Centro Médico Nacional de Occidente, Instituto Mexicano de Seguro Social, Guadalajara, Jalisco, México
| | - José de Jesús Guerrero-García
- Banco de Sangre Central, Unidad Médica de Alta Especialidad, Hospital de Especialidades, Centro Médico Nacional de Occidente, Instituto Mexicano de Seguro Social, Guadalajara, Jalisco, México
| | - Daniel Ortuño-Sahagún
- Laboratorio de Neuroinmunobiología Molecular, Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Jalisco, México.
| |
Collapse
|
36
|
Wang W, Wang C, Chen W, Ding S. Advances in immunological research of amphioxus. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 118:103992. [PMID: 33387559 DOI: 10.1016/j.dci.2020.103992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 12/17/2020] [Accepted: 12/26/2020] [Indexed: 06/12/2023]
Abstract
Amphioxus, one of the most closely related invertebrates to vertebrates, is an important animal model for studying the origin and evolution of vertebrate immunity, especially the transition from innate immunity to adaptive immunity. The current research progresses of amphioxus in the field of immune organs, immune cells, complement system, cytokines, nuclear factor kappa B, immune-related lectins and enzymes are summarized, and some issues that remain to be understood or are in need of further clarification are highlighted. We hope to provide references for more in-depth study of the amphioxus immune system and lay a solid foundation for the construction of three-dimensional immune network in amphioxus from ontogeny to phylogeny.
Collapse
Affiliation(s)
- Wenjun Wang
- School of Life Sciences, Ludong University, Yantai, 264025, People's Republic of China
| | - Changliu Wang
- School of Life Sciences, Ludong University, Yantai, 264025, People's Republic of China.
| | - Wei Chen
- School of Life Sciences, Ludong University, Yantai, 264025, People's Republic of China; Yantai Productivity Promotion Center, Yantai, 264003, People's Republic of China
| | - Shuo Ding
- School of Life Sciences, Ludong University, Yantai, 264025, People's Republic of China
| |
Collapse
|
37
|
Chen M, Li Y, Huang X, Gu Y, Li S, Yin P, Zhang L, Tang P. Skeleton-vasculature chain reaction: a novel insight into the mystery of homeostasis. Bone Res 2021; 9:21. [PMID: 33753717 PMCID: PMC7985324 DOI: 10.1038/s41413-021-00138-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 11/18/2020] [Accepted: 12/16/2020] [Indexed: 02/01/2023] Open
Abstract
Angiogenesis and osteogenesis are coupled. However, the cellular and molecular regulation of these processes remains to be further investigated. Both tissues have recently been recognized as endocrine organs, which has stimulated research interest in the screening and functional identification of novel paracrine factors from both tissues. This review aims to elaborate on the novelty and significance of endocrine regulatory loops between bone and the vasculature. In addition, research progress related to the bone vasculature, vessel-related skeletal diseases, pathological conditions, and angiogenesis-targeted therapeutic strategies are also summarized. With respect to future perspectives, new techniques such as single-cell sequencing, which can be used to show the cellular diversity and plasticity of both tissues, are facilitating progress in this field. Moreover, extracellular vesicle-mediated nuclear acid communication deserves further investigation. In conclusion, a deeper understanding of the cellular and molecular regulation of angiogenesis and osteogenesis coupling may offer an opportunity to identify new therapeutic targets.
Collapse
Affiliation(s)
- Ming Chen
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Yi Li
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Xiang Huang
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Ya Gu
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Shang Li
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Pengbin Yin
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China.
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China.
| | - Licheng Zhang
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China.
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China.
| | - Peifu Tang
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China.
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China.
| |
Collapse
|
38
|
Vanza JD, Patel RB, Patel MR. Nanocarrier centered therapeutic approaches: Recent developments with insight towards the future in the management of lung cancer. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.102070] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
39
|
Cerezo-Wallis D, Contreras-Alcalde M, Troulé K, Catena X, Mucientes C, Calvo TG, Cañón E, Tejedo C, Pennacchi PC, Hogan S, Kölblinger P, Tejero H, Chen AX, Ibarz N, Graña-Castro O, Martinez L, Muñoz J, Ortiz-Romero P, Rodriguez-Peralto JL, Gómez-López G, Al-Shahrour F, Rabadán R, Levesque MP, Olmeda D, Soengas MS. Midkine rewires the melanoma microenvironment toward a tolerogenic and immune-resistant state. Nat Med 2020; 26:1865-1877. [PMID: 33077955 DOI: 10.1038/s41591-020-1073-3] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 08/20/2020] [Indexed: 12/14/2022]
Abstract
An open question in aggressive cancers such as melanoma is how malignant cells can shift the immune system to pro-tumorigenic functions. Here we identify midkine (MDK) as a melanoma-secreted driver of an inflamed, but immune evasive, microenvironment that defines poor patient prognosis and resistance to immune checkpoint blockade. Mechanistically, MDK was found to control the transcriptome of melanoma cells, allowing for coordinated activation of nuclear factor-κB and downregulation of interferon-associated pathways. The resulting MDK-modulated secretome educated macrophages towards tolerant phenotypes that promoted CD8+ T cell dysfunction. In contrast, genetic targeting of MDK sensitized melanoma cells to anti-PD-1/anti-PD-L1 treatment. Emphasizing the translational relevance of these findings, the expression profile of MDK-depleted tumors was enriched in key indicators of a good response to immune checkpoint blockers in independent patient cohorts. Together, these data reveal that MDK acts as an internal modulator of autocrine and paracrine signals that maintain immune suppression in aggressive melanomas.
Collapse
Affiliation(s)
- Daniela Cerezo-Wallis
- Melanoma Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Marta Contreras-Alcalde
- Melanoma Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Kevin Troulé
- Bioinformatics Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Xavier Catena
- Melanoma Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Cynthia Mucientes
- Melanoma Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Tonantzin G Calvo
- Melanoma Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Estela Cañón
- Melanoma Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Cristina Tejedo
- Melanoma Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Paula C Pennacchi
- Melanoma Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Sabrina Hogan
- Department of Dermatology, University of Zurich Hospital, Zurich, Switzerland
| | - Peter Kölblinger
- Department of Dermatology, University of Zurich Hospital, Zurich, Switzerland
| | - Héctor Tejero
- Bioinformatics Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Andrew X Chen
- Program for Mathematical Genomics, Departament of Systems Biology, Departament of Biomedical Informatics, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Nuria Ibarz
- Proteomics Unit, Biotechnology Programme, Spanish National Cancer Research Centre (CNIO) and ProteoRed-ISCIII, Madrid, Madrid, Spain
| | - Osvaldo Graña-Castro
- Bioinformatics Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Lola Martinez
- Proteomics Unit, Biotechnology Programme, Spanish National Cancer Research Centre (CNIO) and ProteoRed-ISCIII, Madrid, Madrid, Spain
| | - Javier Muñoz
- Flow Cytometry Unit, Biotechnology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Madrid, Spain
| | - Pablo Ortiz-Romero
- Dermatology Service, Hospital 12 de Octubre, Universidad Complutense Madrid Medical School, Madrid, Spain
| | - José L Rodriguez-Peralto
- Instituto de Investigación i+12, Hospital 12 de Octubre, Universidad Complutense Madrid Medical School, Madrid, Spain.,Pathology Service, Hospital 12 de Octubre, Universidad Complutense Madrid Medical School, Madrid, Spain
| | - Gonzalo Gómez-López
- Bioinformatics Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Fátima Al-Shahrour
- Bioinformatics Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Raúl Rabadán
- Program for Mathematical Genomics, Departament of Systems Biology, Departament of Biomedical Informatics, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Mitchell P Levesque
- Department of Dermatology, University of Zurich Hospital, Zurich, Switzerland
| | - David Olmeda
- Melanoma Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain.
| | - María S Soengas
- Melanoma Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain.
| |
Collapse
|
40
|
Midkine-a functions as a universal regulator of proliferation during epimorphic regeneration in adult zebrafish. PLoS One 2020; 15:e0232308. [PMID: 32530962 PMCID: PMC7292404 DOI: 10.1371/journal.pone.0232308] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 05/28/2020] [Indexed: 12/20/2022] Open
Abstract
Zebrafish have the ability to regenerate damaged cells and tissues by activating quiescent stem and progenitor cells or reprogramming differentiated cells into regeneration-competent precursors. Proliferation among the cells that will functionally restore injured tissues is a fundamental biological process underlying regeneration. Midkine-a is a cytokine growth factor, whose expression is strongly induced by injury in a variety of tissues across a range of vertebrate classes. Using a zebrafish Midkine-a loss of function mutant, we evaluated regeneration of caudal fin, extraocular muscle and retinal neurons to investigate the function of Midkine-a during epimorphic regeneration. In wildtype zebrafish, injury among these tissues induces robust proliferation and rapid regeneration. In Midkine-a mutants, the initial proliferation in each of these tissues is significantly diminished or absent. Regeneration of the caudal fin and extraocular muscle is delayed; regeneration of the retina is nearly completely absent. These data demonstrate that Midkine-a is universally required in the signaling pathways that convert tissue injury into the initial burst of cell proliferation. Further, these data highlight differences in the molecular mechanisms that regulate epimorphic regeneration in zebrafish.
Collapse
|
41
|
Hayward S, Gachehiladze M, Badr N, Andrijes R, Molostvov G, Paniushkina L, Sopikova B, Slobodová Z, Mgebrishvili G, Sharma N, Horimoto Y, Burg D, Robertson G, Hanby A, Hoar F, Rea D, Eckhardt BL, Ueno NT, Nazarenko I, Long HM, van Laere S, Shaaban AM, Berditchevski F. The CD151-midkine pathway regulates the immune microenvironment in inflammatory breast cancer. J Pathol 2020; 251:63-73. [PMID: 32129471 DOI: 10.1002/path.5415] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 01/27/2020] [Accepted: 02/25/2020] [Indexed: 12/19/2022]
Abstract
The immune microenvironment in inflammatory breast cancer (IBC) is poorly characterised, and molecular and cellular pathways that control accumulation of various immune cells in IBC tissues remain largely unknown. Here, we discovered a novel pathway linking the expression of the tetraspanin protein CD151 in tumour cells with increased accumulation of macrophages in cancerous tissues. It is notable that elevated expression of CD151 and a higher number of tumour-infiltrating macrophages correlated with better patient responses to chemotherapy. Accordingly, CD151-expressing IBC xenografts were characterised by the increased infiltration of macrophages. In vitro migration experiments demonstrated that CD151 stimulates the chemoattractive potential of IBC cells for monocytes via mechanisms involving midkine (a heparin-binding growth factor), integrin α6β1, and production of extracellular vesicles (EVs). Profiling of chemokines secreted by IBC cells demonstrated that CD151 increases production of midkine. Purified midkine specifically stimulated migration of monocytes, but not other immune cells. Further experiments demonstrated that the chemoattractive potential of IBC-derived EVs is blocked by anti-midkine antibodies. These results demonstrate for the first time that changes in the expression of a tetraspanin protein by tumour cells can affect the formation of the immune microenvironment by modulating recruitment of effector cells to cancerous tissues. Therefore, a CD151-midkine pathway can be considered as a novel target for controlled changes of the immune landscape in IBC. © 2020 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Steven Hayward
- Institute of Cancer and Genomic Sciences, The University of Birmingham, Birmingham, UK
| | - Mariam Gachehiladze
- Department of Clinical and Molecular Pathology, Palacký Univerzity, Olomouc, Czech Republic
| | - Nahla Badr
- Institute of Cancer and Genomic Sciences, The University of Birmingham, Birmingham, UK.,Department of Pathology, Menoufia University School of Medicine, Menoufia, Egypt
| | - Regina Andrijes
- Institute of Cancer and Genomic Sciences, The University of Birmingham, Birmingham, UK
| | - Guerman Molostvov
- Institute of Cancer and Genomic Sciences, The University of Birmingham, Birmingham, UK
| | - Liliia Paniushkina
- Faculty of Medicine, Institute for Infection Prevention and Hospital Epidemiology, Medical Center - University of Freiburg, Freiburg, Germany
| | - Barbora Sopikova
- Department of Clinical and Molecular Pathology, Palacký Univerzity, Olomouc, Czech Republic
| | - Zuzana Slobodová
- Department of Clinical and Molecular Pathology, Palacký Univerzity, Olomouc, Czech Republic
| | - Giorgi Mgebrishvili
- Department of Clinical and Molecular Pathology, Palacký Univerzity, Olomouc, Czech Republic
| | - Nisha Sharma
- Breast Unit, St James Hospital, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Yoshiya Horimoto
- Department of Breast Surgical Oncology, Juntendo University School of Medicine, Tokyo, Japan
| | | | | | - Andrew Hanby
- University of Leeds, Leeds Institute of Cancer and Pathology (LICAP) Leeds, Leeds, UK
| | - Fiona Hoar
- Hospital, Sandwell and West Birmingham Hospitals, Department of General and Breast Surgery, Birmingham, UK
| | - Daniel Rea
- Institute of Cancer and Genomic Sciences, The University of Birmingham, Birmingham, UK
| | - Bedrich L Eckhardt
- Olivia Newton-John Cancer Research Institute, Heidelberg, Australia.,Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, Houston, TX, USA
| | - Naoto T Ueno
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, Houston, TX, USA
| | - Irina Nazarenko
- Faculty of Medicine, Institute for Infection Prevention and Hospital Epidemiology, Medical Center - University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Heather M Long
- Institute of Cancer and Genomic Sciences, The University of Birmingham, Birmingham, UK
| | - Steven van Laere
- Translational Cancer Research Unit Center for Oncological Research, University Antwerp, Antwerp, Belgium
| | - Abeer M Shaaban
- Institute of Cancer and Genomic Sciences, The University of Birmingham, Birmingham, UK
| | - Fedor Berditchevski
- Institute of Cancer and Genomic Sciences, The University of Birmingham, Birmingham, UK
| |
Collapse
|
42
|
Abstract
Pleiotrophin (PTN) is a potent mitogenic cytokine with a high affinity for the polysaccharide glycosaminoglycan (GAG). Although it is most strongly associated with neural development during embryogenesis and the neonatal period, its expression has also been linked to a plethora of other physiological events including cancer metastasis, angiogenesis, bone development, and inflammation. A considerable amount of research has been carried out to understand the mechanisms by which PTN regulates these events. In particular, PTN has now been shown to bind a diverse collection of receptors including many GAG-containing proteoglycans. These interactions lead to the activation of many intracellular kinases and, ultimately, activation and transformation of cells. Structural studies of PTN in complex with both GAG and domains from its non-proteoglycan receptors reveal a binding mechanism that relies on electrostatic interactions and points to PTN-induced receptor oligomerization as one of the possible ways PTN uses to control cellular functions.
Collapse
|
43
|
Wang P, Mao YM, Zhao CN, Wang JB, Li XM, Ye DQ, Pan HF. Association of Midkine and Pleiotrophin Gene Polymorphisms With Systemic Lupus Erythematosus Susceptibility in Chinese Han Population. Front Immunol 2020; 11:110. [PMID: 32153561 PMCID: PMC7046794 DOI: 10.3389/fimmu.2020.00110] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 01/15/2020] [Indexed: 02/04/2023] Open
Abstract
In a previous study, we have reported an increased plasma midkine (MK) and pleiotrophin (PTN) concentrations in patients with systemic lupus erythematosus (SLE) and the increase in MK and PTN associated with inflammatory cytokines interleukin (IL)-17 level and some clinical manifestations, suggesting the underlying association of MK and PTN with SLE. This study was conducted to investigate the association between common single-nucleotide polymorphisms (SNPs) in the MK and PTN gene and SLE susceptibility. A total of 989 subjects (496 SLE patients and 493 healthy controls) were included and genotyped for three MK SNPs and seven PTN SNPs in using improved multiple ligase detection reaction (iMLDR). Results have demonstrated no significant differences for genotype and allele frequencies in all 10 SNPs between SLE patients and healthy controls. Case-only analysis in SLE revealed that, in MK gene, the genotype frequency of AA/AG (rs35324223) was significantly lower in patients with photosensitivity than those without; the allele frequency of A/G (rs20542) was significantly higher in patients without serositis. In PTN gene, the A/G allele frequency (rs322236), C/T allele frequency, and TT/CT genotype frequency (rs6970141) showed significantly increased results in patients with immunological disorder compared to those without. Furthermore, no significant differences in plasma MK and PTN concentrations with its SNPs genotypes were found. MK and PTN SNPs showed no associations with SLE genetic susceptibility, but it may be associated with the course of this disease; further studies are needed to focus on the mechanism of MK and PTN genes in the pathogenesis of SLE.
Collapse
Affiliation(s)
- Peng Wang
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Yan-Mei Mao
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Hefei, China
| | - Chan-Na Zhao
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Hefei, China
| | - Jie-Bing Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Hefei, China
| | - Xiao-Mei Li
- Department of Rheumatology and Immunology, Anhui Provincial Hospital, Hefei, China
| | - Dong-Qing Ye
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Hefei, China
| | - Hai-Feng Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Major Autoimmune Diseases, Hefei, China
| |
Collapse
|
44
|
Tsai SL, Baselga-Garriga C, Melton DA. Midkine is a dual regulator of wound epidermis development and inflammation during the initiation of limb regeneration. eLife 2020; 9:50765. [PMID: 31934849 PMCID: PMC6959999 DOI: 10.7554/elife.50765] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 12/23/2019] [Indexed: 12/13/2022] Open
Abstract
Formation of a specialized wound epidermis is required to initiate salamander limb regeneration. Yet little is known about the roles of the early wound epidermis during the initiation of regeneration and the mechanisms governing its development into the apical epithelial cap (AEC), a signaling structure necessary for outgrowth and patterning of the regenerate. Here, we elucidate the functions of the early wound epidermis, and further reveal midkine (mk) as a dual regulator of both AEC development and inflammation during the initiation of axolotl limb regeneration. Through loss- and gain-of-function experiments, we demonstrate that mk acts as both a critical survival signal to control the expansion and function of the early wound epidermis and an anti-inflammatory cytokine to resolve early injury-induced inflammation. Altogether, these findings unveil one of the first identified regulators of AEC development and provide fundamental insights into early wound epidermis function, development, and the initiation of limb regeneration.
Collapse
Affiliation(s)
- Stephanie L Tsai
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, United States.,Department of Molecular and Cellular Biology, Harvard University, Cambridge, United States
| | - Clara Baselga-Garriga
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, United States.,Department of Molecular and Cellular Biology, Harvard University, Cambridge, United States
| | - Douglas A Melton
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, United States
| |
Collapse
|
45
|
Midkine-a Is Required for Cell Cycle Progression of Müller Glia during Neuronal Regeneration in the Vertebrate Retina. J Neurosci 2019; 40:1232-1247. [PMID: 31882403 PMCID: PMC7002140 DOI: 10.1523/jneurosci.1675-19.2019] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 11/27/2019] [Accepted: 12/17/2019] [Indexed: 12/25/2022] Open
Abstract
In the retina of zebrafish, Müller glia have the ability to reprogram into stem cells capable of regenerating all classes of retinal neurons and restoring visual function. Understanding the cellular and molecular mechanisms controlling the stem cell properties of Müller glia in zebrafish may provide cues to unlock the regenerative potential in the mammalian nervous system. Midkine is a cytokine/growth factor with multiple roles in neural development, tissue repair, and disease. In the retina of zebrafish, Müller glia have the ability to reprogram into stem cells capable of regenerating all classes of retinal neurons and restoring visual function. Understanding the cellular and molecular mechanisms controlling the stem cell properties of Müller glia in zebrafish may provide cues to unlock the regenerative potential in the mammalian nervous system. Midkine is a cytokine/growth factor with multiple roles in neural development, tissue repair, and disease. In midkine-a loss-of-function mutants of both sexes, Müller glia initiate the appropriate reprogramming response to photoreceptor death by increasing expression of stem cell-associated genes, and entering the G1 phase of the cell cycle. However, transition from G1 to S phase is blocked in the absence of Midkine-a, resulting in significantly reduced proliferation and selective failure to regenerate cone photoreceptors. Failing to progress through the cell cycle, Müller glia undergo reactive gliosis, a pathological hallmark in the injured CNS of mammals. Finally, we determined that the Midkine-a receptor, anaplastic lymphoma kinase, is upstream of the HLH regulatory protein, Id2a, and of the retinoblastoma gene, p130, which regulates progression through the cell cycle. These results demonstrate that Midkine-a functions as a core component of the mechanisms that regulate proliferation of stem cells in the injured CNS. SIGNIFICANCE STATEMENT The death of retinal neurons and photoreceptors is a leading cause of vision loss. Regenerating retinal neurons is a therapeutic goal. Zebrafish can regenerate retinal neurons from intrinsic stem cells, Müller glia, and are a powerful model to understand how stem cells might be used therapeutically. Midkine-a, an injury-induced growth factor/cytokine that is expressed by Müller glia following neuronal death, is required for Müller glia to progress through the cell cycle. The absence of Midkine-a suspends proliferation and neuronal regeneration. With cell cycle progression stalled, Müller glia undergo reactive gliosis, a pathological hallmark of the mammalian retina. This work provides a unique insight into mechanisms that control the cell cycle during neuronal regeneration.
Collapse
|
46
|
Gautron J, Guyot N, Brionne A, Réhault-Godbert S. Bioactive Minor Egg Components. EGGS AS FUNCTIONAL FOODS AND NUTRACEUTICALS FOR HUMAN HEALTH 2019. [DOI: 10.1039/9781788013833-00259] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
In the last 15 years, the development of functional genomics has increased the number of egg proteins identified from 50 to about 1300. These proteins are initially present in eggs to support a harmonious embryonic development. Consequently, this closed embryonic chamber contains molecules exhibiting diverse functions, including defense, nutrition and many predicted biological activities, which have been investigated using both bioinformatics and experimental investigations. In this chapter, we focus on some very interesting activities of high potential reported for minor egg proteins (excluding ovalbumin, ovotransferrin and lysozyme). The shell matrix proteins are involved in the calcification process to define and control the final texture of the shell and thereby its mechanical properties. Antimicrobial proteins are part of innate immunity and are mainly present in the white and vitelline membranes. They encompass several protein families, including protease inhibitors, vitamin-binding proteins, defensins, LBP-PLUNC family proteins and heparin-binding proteins. The egg also possesses additional bioactive proteins with direct anti-cancerous and antioxidant activities or whose biochemical properties are currently used to develop diagnostic tools and strategies for targeted therapy. Finally, this chapter also reports some emerging functions in tissue remodeling/wound healing and proposes some relevant bioactive candidates and research fields that would be interesting to investigate further.
Collapse
Affiliation(s)
- J. Gautron
- INRA, BOA, Université de Tours 37380 Nouzilly France
| | - N. Guyot
- INRA, BOA, Université de Tours 37380 Nouzilly France
| | - A. Brionne
- INRA, BOA, Université de Tours 37380 Nouzilly France
| | | |
Collapse
|
47
|
Herradon G, Ramos-Alvarez MP, Gramage E. Connecting Metainflammation and Neuroinflammation Through the PTN-MK-RPTPβ/ζ Axis: Relevance in Therapeutic Development. Front Pharmacol 2019; 10:377. [PMID: 31031625 PMCID: PMC6474308 DOI: 10.3389/fphar.2019.00377] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 03/26/2019] [Indexed: 12/15/2022] Open
Abstract
Inflammation is a common factor of pathologies such as obesity, type 2 diabetes or neurodegenerative diseases. Chronic inflammation is considered part of the pathogenic mechanisms of different disorders associated with aging. Interestingly, peripheral inflammation and the associated metabolic alterations not only facilitate insulin resistance and diabetes but also neurodegenerative disorders. Therefore, the identification of novel pathways, common to the development of these diseases, which modulate the immune response and signaling is key. It will provide highly relevant information to advance our knowledge of the multifactorial process of aging, and to establish new biomarkers and/or therapeutic targets to counteract the underlying chronic inflammatory processes. One novel pathway that regulates peripheral and central immune responses is triggered by the cytokines pleiotrophin (PTN) and midkine (MK), which bind its receptor, Receptor Protein Tyrosine Phosphatase (RPTP) β/ζ, and inactivate its phosphatase activity. In this review, we compile a growing body of knowledge suggesting that PTN and MK modulate the immune response and/or inflammation in different pathologies characterized by peripheral inflammation associated with insulin resistance, such as aging, and in central disorders characterized by overt neuroinflammation, such as neurodegenerative diseases and endotoxemia. Evidence strongly suggests that regulation of the PTN and MK signaling pathways may provide new therapeutic opportunities particularly in those neurological disorders characterized by increased PTN and/or MK cerebral levels and neuroinflammation. Importantly, we discuss existing therapeutics, and others being developed, that modulate these signaling pathways, and their potential use in pathologies characterized by overt neuroinflammation.
Collapse
Affiliation(s)
- Gonzalo Herradon
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - M Pilar Ramos-Alvarez
- Departmento de Química y Bioquímica, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Esther Gramage
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| |
Collapse
|
48
|
Réhault-Godbert S, Guyot N, Nys Y. The Golden Egg: Nutritional Value, Bioactivities, and Emerging Benefits for Human Health. Nutrients 2019; 11:E684. [PMID: 30909449 PMCID: PMC6470839 DOI: 10.3390/nu11030684] [Citation(s) in RCA: 200] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 03/15/2019] [Accepted: 03/19/2019] [Indexed: 12/26/2022] Open
Abstract
Egg is an encapsulated source of macro and micronutrients that meet all requirements to support embryonic development until hatching. The perfect balance and diversity in its nutrients along with its high digestibility and its affordable price has put the egg in the spotlight as a basic food for humans. However, egg still has to face many years of nutritionist recommendations aiming at restricting egg consumption to limit cardiovascular diseases incidence. Most experimental, clinical, and epidemiologic studies concluded that there was no evidence of a correlation between dietary cholesterol brought by eggs and an increase in plasma total-cholesterol. Egg remains a food product of high nutritional quality for adults including elderly people and children and is extensively consumed worldwide. In parallel, there is compelling evidence that egg also contains many and still-unexplored bioactive compounds, which may be of high interest in preventing/curing diseases. This review will give an overview of (1) the main nutritional characteristics of chicken egg, (2) emerging data related to egg bioactive compounds, and (3) some factors affecting egg composition including a comparison of nutritional value between eggs from various domestic species.
Collapse
Affiliation(s)
| | - Nicolas Guyot
- Biologie des Oiseaux et Aviculture, INRA, Université de Tours, 37380 Nouzilly, France.
| | - Yves Nys
- Biologie des Oiseaux et Aviculture, INRA, Université de Tours, 37380 Nouzilly, France.
| |
Collapse
|
49
|
Sorrelle N, Ganguly D, Dominguez ATA, Zhang Y, Huang H, Dahal LN, Burton N, Ziemys A, Brekken RA. Improved Multiplex Immunohistochemistry for Immune Microenvironment Evaluation of Mouse Formalin-Fixed, Paraffin-Embedded Tissues. THE JOURNAL OF IMMUNOLOGY 2018; 202:292-299. [PMID: 30510069 DOI: 10.4049/jimmunol.1800878] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 10/29/2018] [Indexed: 01/12/2023]
Abstract
Immune profiling of tissue through multiplex immunohistochemistry is important for the investigation of immune cell dynamics, and it can contribute to disease prognosis and evaluation of treatment response in cancer patients. However, protocols for mouse formalin-fixed, paraffin-embedded tissue have been less successful. Given that formalin fixation and paraffin embedding remains the most common preparation method for processing mouse tissue, this has limited the options to study the immune system and the impact of novel therapeutics in preclinical models. In an attempt to address this, we developed an improved immunohistochemistry protocol with a more effective Ag-retrieval buffer. We also validated 22 Abs specific for mouse immune cell markers to distinguish B cells, T cells, NK cells, macrophages, dendritic cells, and neutrophils. In addition, we designed and tested novel strategies to identify immune cells for which unique Abs are currently not available. Last, in the 4T1 model of breast cancer, we demonstrate the utility of our protocol and Ab panels in the quantitation and spatial distribution of immune cells.
Collapse
Affiliation(s)
- Noah Sorrelle
- Division of Surgical Oncology, Department of Surgery, Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Debolina Ganguly
- Division of Surgical Oncology, Department of Surgery, Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Adrian T A Dominguez
- Division of Surgical Oncology, Department of Surgery, Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Yuqing Zhang
- Division of Surgical Oncology, Department of Surgery, Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Huocong Huang
- Division of Surgical Oncology, Department of Surgery, Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Lekh N Dahal
- Centre for Cancer Immunology, Faculty of Medicine, Southampton General Hospital, University of Southampton, Southampton 016 6YD, United Kingdom
| | - Natalie Burton
- Division of Surgical Oncology, Department of Surgery, Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Arturas Ziemys
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030; and
| | - Rolf A Brekken
- Division of Surgical Oncology, Department of Surgery, Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX 75390; .,Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| |
Collapse
|
50
|
Gao Z, Qu B, Yao L, Ma Z, Cui P, Zhang S. Identification and functional characterization of amphioxus Miple, ancestral type of vertebrate midkine/pleiotrophin homologues. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2018; 89:31-43. [PMID: 30096337 DOI: 10.1016/j.dci.2018.08.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 08/06/2018] [Accepted: 08/06/2018] [Indexed: 06/08/2023]
Abstract
Midkine (MK) and pleiotrophin (PTN) are the only two members of heparin-binding growth factor family. MK/PTN homologues found from Drosophila to humans are shown to have antibacterial activities and their antibacterial domains are conserved during evolution. However, little is known about MK/PTN homologue in the basal chordate amphioxus, and overall, information regarding MK/PTN homologues is rather limited in invertebrates. In this study, we identified a single MK/PTN homologue in Branchiostoma japonicum, termed BjMiple, which has a novel domain structure of PTN-PTNr1-PTNr2, and represents the ancestral form of vertebrate MK/PTN family proteins. BjMiple was expressed mainly in the ovary in a tissue-dependent fashion, and its expression was remarkably up-regulated following challenge with bacteria or their signature molecules LPS and LTA, suggesting its involvement in antibacterial responses. Functional assays revealed that BjMiple had strong antimicrobial activity, capable of killing a panel of Gram-negative and Gram-positive bacteria via a membranolytic mechanism, including interaction with bacterial membrane via LPS and LTA, membrane depolarization and high intracellular levels of ROS. Importantly, strong antibacterial activity was localized in PTN42-61 and PTNr142-66. Additionally, BjMiple and its derived peptides PTN42-61 and PTNr142-66 were not cytotoxic to human RBCs and mammalian cells. Taken together, our study suggests that amphioxus Miple is the ancestral type of vertebrate MK/PTN family homologues, and can play important roles as innate peptide antibiotics, which renders it a promising template for the design of novel peptide antibiotics against multi-drug resistant bacteria.
Collapse
Affiliation(s)
- Zhan Gao
- Laboratory for Evolution & Development, Institute of Evolution & Marine Biodiversity, Qingdao, 266003, China; Department of Marine Biology, Ocean University of China, Qingdao, 266003, China
| | - Baozhen Qu
- Laboratory for Evolution & Development, Institute of Evolution & Marine Biodiversity, Qingdao, 266003, China; Department of Marine Biology, Ocean University of China, Qingdao, 266003, China
| | - Lan Yao
- Laboratory for Evolution & Development, Institute of Evolution & Marine Biodiversity, Qingdao, 266003, China; Department of Marine Biology, Ocean University of China, Qingdao, 266003, China
| | - Zengyu Ma
- Laboratory for Evolution & Development, Institute of Evolution & Marine Biodiversity, Qingdao, 266003, China; Department of Marine Biology, Ocean University of China, Qingdao, 266003, China
| | - Pengfei Cui
- Department of Marine Biology, Ocean University of China, Qingdao, 266003, China
| | - Shicui Zhang
- Laboratory for Evolution & Development, Institute of Evolution & Marine Biodiversity, Qingdao, 266003, China; Department of Marine Biology, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China.
| |
Collapse
|