1
|
Jacobsen DP, Fjeldstad HE, Olsen MB, Sugulle M, Staff AC. Microchimerism and pregnancy complications with placental dysfunction. Semin Immunopathol 2025; 47:21. [PMID: 40067448 PMCID: PMC11897092 DOI: 10.1007/s00281-025-01045-w] [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: 04/12/2024] [Accepted: 01/27/2025] [Indexed: 03/15/2025]
Abstract
Cells cross the placenta during pregnancy, resulting in proliferation of semiallogeneic cells in the mother and fetus decades later. This phenomenon, termed microchimerism, is documented across mammalian species, implying an evolutionary benefit. Still, short- and long-term effects remain uncertain. Here, we review the dynamics of microchimerism of fetal, maternal, and mother of the proband origin in relation to increasing gestational age and pregnancy complications associated with placental dysfunction including preeclampsia, fetal growth restriction, preterm labor, recurrent miscarriage, and diabetes. We use the two-stage model of preeclampsia as a framework. We recently published a series of papers independently linking increased fetal microchimerism to markers of placental dysfunction (stage 1), severe maternal hypertension (stage 2) and poor glucose control. Placental dysfunction may influence the intrinsic properties of fetal stem cells. Mesenchymal and hematopoietic stem cells isolated from cord blood during preeclampsia display reduced proliferative potential in vitro. Moreover, preeclampsia is shown to disrupt paracrine signaling in mesenchymal stem cells of the umbilical cord. Undesired properties in cells transferred to the mother could have profound negative effects on maternal health. Finally, recent studies indicate that microchimerism is involved in inducing maternal-fetal tolerance. Disruption of this process is associated with pregnancy complications. Long term, the persistence of microchimerism is necessary to sustain specific regulatory T cell populations in mice. This likely plays a role in the proband's future pregnancies and long-term maternal and offspring health. Current evidence indicates that advancements in our understanding of microchimerism could be instrumental in promoting reproductive and long-term health.
Collapse
Affiliation(s)
- Daniel Pitz Jacobsen
- Faculty of Medicine, University of Oslo, Oslo, Norway.
- Division of Obstetrics and Gynaecology, Oslo University Hospital, Oslo, Norway.
- Division of Obstetrics and Gynaecology, Oslo University Hospital, Oslo University Hospital, Kirkeveien 166, Box 4956, Oslo, Nydalen, Oslo, 0450, 0424, PO, Norway.
| | - Heidi E Fjeldstad
- Faculty of Medicine, University of Oslo, Oslo, Norway
- Division of Obstetrics and Gynaecology, Oslo University Hospital, Oslo, Norway
| | - Maria B Olsen
- Faculty of Medicine, University of Oslo, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - Meryam Sugulle
- Faculty of Medicine, University of Oslo, Oslo, Norway
- Division of Obstetrics and Gynaecology, Oslo University Hospital, Oslo, Norway
| | - Anne Cathrine Staff
- Faculty of Medicine, University of Oslo, Oslo, Norway
- Division of Obstetrics and Gynaecology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
2
|
Bergmann L, Afflerbach AK, Yuan T, Pantel K, Smit DJ. Lessons (to be) learned from liquid biopsies: assessment of circulating cells and cell-free DNA in cancer and pregnancy-acquired microchimerism. Semin Immunopathol 2025; 47:14. [PMID: 39893314 PMCID: PMC11787191 DOI: 10.1007/s00281-025-01042-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: 08/05/2024] [Accepted: 01/20/2025] [Indexed: 02/04/2025]
Abstract
Tumors constantly shed cancer cells that are considered the mediators of metastasis via the blood stream. Analysis of circulating cells and circulating cell-free DNA (cfDNA) in liquid biopsies, mostly taken from peripheral blood, have emerged as powerful biomarkers in oncology, as they enable the detection of genomic aberrations. Similarly, liquid biopsies taken from pregnant women serve as prenatal screening test for an abnormal number of chromosomes in the fetus, e.g., via the analysis of microchimeric fetal cells and cfDNA circulating in maternal blood. Liquid biopsies are minimally invasive and, consequently, associated with reduced risks for the patients. However, different challenges arise in oncology and pregnancy-acquired liquid biopsies with regard to the analyte concentration and biological (background) noise among other factors. In this review, we highlight the unique biological properties of circulating tumor cells (CTC), summarize the various techniques that have been developed for the enrichment, detection and analysis of CTCs as well as for analysis of genetic and epigenetic aberrations in cfDNA and highlight the range of possible clinical applications. Lastly, the potential, but also the challenges of liquid biopsies in oncology as well as their translational value for the analysis of pregnancy-acquired microchimerism are discussed.
Collapse
Affiliation(s)
- Lina Bergmann
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, Hamburg, 20246, Germany
| | - Ann-Kristin Afflerbach
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, Hamburg, 20246, Germany
| | - Tingjie Yuan
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, Hamburg, 20246, Germany
| | - Klaus Pantel
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, Hamburg, 20246, Germany.
| | - Daniel J Smit
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, Hamburg, 20246, Germany.
| |
Collapse
|
3
|
Slaats E, Bramreiter B, Chua KJ, Quilang RC, Sallinger K, Eikmans M, Kroneis T. Maternal microchimeric cell trafficking and its biological consequences depend on the onset of inflammation at the feto-maternal interface. Semin Immunopathol 2025; 47:8. [PMID: 39820729 PMCID: PMC11742462 DOI: 10.1007/s00281-025-01037-w] [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: 05/13/2024] [Accepted: 01/06/2025] [Indexed: 01/19/2025]
Abstract
Microchimerism is defined as the presence of a small population of genetically distinct cells within a host that is derived from another individual. Throughout pregnancy, maternal and fetal cells are known to traffic across the feto-maternal interface and result in maternal and fetal microchimerism, respectively. However, the routes of cell transfer, the molecular signaling as well as the timing in which trafficking takes place are still not completely understood. Recently, the presence of inflammation at the feto-maternal interface has been linked with maternal microchimeric cells modulating organ development in the fetus. Here, we review the current literature and suggest that inflammatory processes at the feto-maternal interface tissues are a physiological prerequisite for the establishment of microchimerism. We further propose a spatio-temporal corridor of microchimeric cell migration to potentially explain some biological effects of microchimerism. Additionally, we elaborate on the possible consequences of a shift in this spatio-temporal corridor, potentially responsible for the development of pathologies in the neonate.
Collapse
Affiliation(s)
- Emiel Slaats
- Gottfried Schatz Research Center, Division of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
| | - Bernadette Bramreiter
- Gottfried Schatz Research Center, Division of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
| | - Kristine J Chua
- Department of Anthropology, University of California Santa Barbara, Santa Barbara, CA, USA
- Department of Anthropology, University of Notre Dame, Notre Dame, CA, USA
| | - Rachel C Quilang
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
| | - Katja Sallinger
- Gottfried Schatz Research Center, Division of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
| | - Michael Eikmans
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
| | - Thomas Kroneis
- Gottfried Schatz Research Center, Division of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria.
| |
Collapse
|
4
|
Kruchen A, Fehse B, Müller I. Clinical relevance of feto-maternal microchimerism in (hematopoietic stem cell) transplantation. Semin Immunopathol 2024; 47:4. [PMID: 39644358 PMCID: PMC11625077 DOI: 10.1007/s00281-024-01028-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 11/14/2024] [Indexed: 12/09/2024]
Abstract
Toleration of a semi-allogeneic fetus in the mother's uterus as well as tolerance after allogeneic hematopoietic stem cell transplantation (HSCT) appear to share some immunologic concepts. The existence of microchimeric cells, and the original idea of a bidirectional cell trafficking between mother and child during pregnancy have been known for decades. Today, origins and mechanisms of persistence of microchimeric cells are intensively being elucidated. Both, the translation of the phenomenon of feto-maternal immune tolerance to donor choice or prevention of graft-versus-host disease (GvHD) in HSCT, and the implications of microchimeric cells in and for HSCT are highly intriguing. Yet, differences in detection methods of microchimeric cells, as well as in transplantation protocols impede the comparison of larger cohorts, and limit potential clinical advice. Still, matching of non-inherited maternal antigens (NIMA), which are expressed on maternal microchimeric cells, demonstrated a strong association with decreased risk for the development of acute GvHD in the context of various transplantation strategies. Despite the fact that advances in graft manipulation and immunosuppression ameliorated the safety and outcome after HSCT, NIMA-matching retained a beneficial role in selection of sibling, child, or maternal donors, as well as for cord blood units. Recent findings indicate the existence of a microchimeric stem cell niche, in which only one dominant microchimeric cell population of only one semi-allogeneic origin persists at a time. This implies that studies regarding the impact of (maternal and fetal) microchimerism (MC) on clinical outcome of HSCT should combine analysis of NIMA and direct detection of microchimeric cells from donor and recipient on the verge of HSCT to be efficiently conclusive.
Collapse
Affiliation(s)
- Anne Kruchen
- Division of Pediatric Stem Cell Transplantation and Immunology, Clinic of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Boris Fehse
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany.
- German Center for Child and Adolescent Health (DZKJ), Hamburg, Germany.
| | - Ingo Müller
- Division of Pediatric Stem Cell Transplantation and Immunology, Clinic of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
- Research Institute Children's Cancer Center Hamburg, 20246, Hamburg, Germany
- German Center for Child and Adolescent Health (DZKJ), Hamburg, Germany
| |
Collapse
|
5
|
Srivastava R, Singh K, Abouhashem AS, Kumar M, Kacar S, Verma SS, Mohanty SK, Sinha M, Ghatak S, Xuan Y, Sen CK. Human fetal dermal fibroblast-myeloid cell diversity is characterized by dominance of pro-healing Annexin1-FPR1 signaling. iScience 2023; 26:107533. [PMID: 37636079 PMCID: PMC10450526 DOI: 10.1016/j.isci.2023.107533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 06/06/2023] [Accepted: 07/28/2023] [Indexed: 08/29/2023] Open
Abstract
Fetal skin achieves scarless wound repair. Dermal fibroblasts play a central role in extracellular matrix deposition and scarring outcomes. Both fetal and gingival wound repair share minimal scarring outcomes. We tested the hypothesis that compared to adult skin fibroblasts, human fetal skin fibroblast diversity is unique and partly overlaps with gingival skin fibroblasts. Human fetal skin (FS, n = 3), gingiva (HGG, n = 13), and mature skin (MS, n = 13) were compared at single-cell resolution. Dermal fibroblasts, the most abundant cluster, were examined to establish a connectome with other skin cells. Annexin1-FPR1 signaling pathway was dominant in both FS as well as HGG fibroblasts and related myeloid cells while scanty in MS fibroblasts. Myeloid-specific FPR1-ORF delivered in murine wound edge using tissue nanotransfection (TNT) technology significantly enhanced the quality of healing. Pseudotime analyses identified the co-existence of an HGG fibroblast subset with FPR1high myeloid cells of fetal origin indicating common underlying biological processes.
Collapse
Affiliation(s)
- Rajneesh Srivastava
- McGowan Institute for Regenerative Medicine, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kanhaiya Singh
- McGowan Institute for Regenerative Medicine, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ahmed S. Abouhashem
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Sharkia Clinical Research Department, Ministry of Health, Zagazig, Egypt
| | - Manishekhar Kumar
- McGowan Institute for Regenerative Medicine, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sedat Kacar
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sumit S. Verma
- McGowan Institute for Regenerative Medicine, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sujit K. Mohanty
- McGowan Institute for Regenerative Medicine, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Mithun Sinha
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Subhadip Ghatak
- McGowan Institute for Regenerative Medicine, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yi Xuan
- McGowan Institute for Regenerative Medicine, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Chandan K. Sen
- McGowan Institute for Regenerative Medicine, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
6
|
Jacobsen DP, Fjeldstad HE, Sugulle M, Johnsen GM, Olsen MB, Kanaan SB, Staff AC. Fetal microchimerism and the two-stage model of preeclampsia. J Reprod Immunol 2023; 159:104124. [PMID: 37541161 DOI: 10.1016/j.jri.2023.104124] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 06/21/2023] [Accepted: 07/25/2023] [Indexed: 08/06/2023]
Abstract
Fetal cells cross the placenta during pregnancy and some have the ability to persist in maternal organs and circulation long-term, a phenomenon termed fetal microchimerism. These cells often belong to stem cell or immune cell lineages. The long-term effects of fetal microchimerism are likely mixed, potentially depending on the amount of fetal cells transferred, fetal-maternal histocompatibility and fetal cell-specific properties. Both human and animal data indicate that fetal-origin cells partake in tissue repair and may benefit maternal health overall. On the other hand, these cells have been implicated in inflammatory diseases by studies showing increased fetal microchimerism in women with autoimmune diseases such as systemic lupus erythematosus and rheumatoid arthritis. During pregnancy, preeclampsia is associated with increased cell-transfer between the mother and fetus, and an increase in immune cell subsets. In the current review, we discuss potential mechanisms of transplacental transfer, including passive leakage across the compromised diffusion barrier and active recruitment of cells residing in the placenta or fetal circulation. Within the conceptual framework of the two-stage model of preeclampsia, where syncytiotrophoblast stress is a common pathophysiological pathway to maternal and fetal clinical features of preeclampsia, we argue that microchimerism may represent a mechanistic link between stage 1 placental dysfunction and stage 2 maternal cardiovascular inflammation and endothelial dysfunction. Finally, we postulate that fetal microchimerism may contribute to the known association between placental syndromes and increased long-term maternal cardiovascular disease risk. Fetal microchimerism research represents an exciting opportunity for developing new disease biomarkers and targeted prophylaxis against maternal diseases.
Collapse
Affiliation(s)
- Daniel P Jacobsen
- Faculty of Medicine, University of Oslo, Oslo, Norway; Division of Obstetrics and Gynaecology, Oslo University Hospital, Ullevål, Oslo, Norway.
| | | | - Meryam Sugulle
- Faculty of Medicine, University of Oslo, Oslo, Norway; Division of Obstetrics and Gynaecology, Oslo University Hospital, Ullevål, Oslo, Norway
| | - Guro M Johnsen
- Division of Obstetrics and Gynaecology, Oslo University Hospital, Ullevål, Oslo, Norway
| | - Maria B Olsen
- Faculty of Medicine, University of Oslo, Oslo, Norway; Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - Sami B Kanaan
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Chimerocyte, Inc., Seattle, WA, USA
| | - Anne Cathrine Staff
- Faculty of Medicine, University of Oslo, Oslo, Norway; Division of Obstetrics and Gynaecology, Oslo University Hospital, Ullevål, Oslo, Norway
| |
Collapse
|
7
|
Alkobtawi M, Sbeih M, Souaid K, Ngô QT, Nassar D, Arbes H, Guillet H, Habibi A, Bartolucci P, Castela M, Aractingi S, Oulès B. Contribution of fetal microchimeric cells to maternal wound healing in sickle cell ulcers. Haematologica 2023; 108:1920-1933. [PMID: 36373248 PMCID: PMC10316260 DOI: 10.3324/haematol.2022.281140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 10/31/2022] [Indexed: 08/18/2024] Open
Abstract
Leg ulcers are a major complication of sickle cell disease (SCD). They are particularly challenging to treat and innovative therapies are needed. We previously showed that the healing of SCD ulcers is delayed because of decreased angiogenesis. During pregnancy, fetal microchimeric cells (FMC) transferred to the mother are recruited to maternal wounds and improve angiogenesis. After delivery, FMC persist in maternal bone marrow for decades. Here, we investigated whether fetal cells could also improve SCD ulcers in the post-partum setting. We found that skin healing was similarly improved in post-partum mice and in pregnant mice, through increased proliferation and angiogenesis. In a SCD mouse model that recapitulates refractory SCD ulcers, we showed that the ulcers of post-partum SCD mice healed more quickly than those of virgin mice. This was associated with the recruitment of fetal cells in maternal wounds where they harbored markers of leukocytes and endothelial cells. In a retrospective cohort of SCD patients, using several parameters we found that SCD women who had ever had a baby had less of a burden related to leg ulcers compared to nulliparous women. Taken together, these results indicate that healing capacities of FMC are maintained long after delivery and may be exploited to promote wound healing in post-partum SCD patients.
Collapse
Affiliation(s)
- Mansour Alkobtawi
- Cutaneous Biology Lab, Institut Cochin, INSERM U1016, UMR 8104, Paris
| | - Maria Sbeih
- Cutaneous Biology Lab, Institut Cochin, INSERM U1016, UMR 8104, Paris
| | - Karim Souaid
- Department of Dermatology, Hôpital Cochin, AP-HP.Centre-Université Paris Cité, Paris, France; University Paris Cité, Faculté de Médecine Paris Centre Santé, Paris
| | - Qui Trung Ngô
- Cutaneous Biology Lab, Institut Cochin, INSERM U1016, UMR 8104, Paris
| | - Dany Nassar
- Cutaneous Biology Lab, Institut Cochin, INSERM U1016, UMR 8104, Paris, France; Department of Dermatology, Hôpital Cochin, AP-HP.Centre-Université Paris Cité, Paris, France; University Paris Cité, Faculté de Médecine Paris Centre Santé, Paris
| | - Hugo Arbes
- Institut de Biologie Intégrative de la Cellule, Genomic structure and Translation Lab, UMR_9198, CEA, CNRS, Université Paris-Saclay, Orsay
| | - Henri Guillet
- Department of Internal Medicine, Red Blood Cell Genetic Diseases Unit, Hôpital Mondor, AP-HP. Hôpitaux Universitaires Henri Mondor, Créteil
| | - Anoosha Habibi
- Department of Internal Medicine, Red Blood Cell Genetic Diseases Unit, Hôpital Mondor, AP-HP. Hôpitaux Universitaires Henri Mondor, Créteil
| | - Pablo Bartolucci
- Department of Internal Medicine, Red Blood Cell Genetic Diseases Unit, Hôpital Mondor, AP-HP. Hôpitaux Universitaires Henri Mondor, Créteil
| | - Mathieu Castela
- Cutaneous Biology Lab, Institut Cochin, INSERM U1016, UMR 8104, Paris
| | - Sélim Aractingi
- Cutaneous Biology Lab, Institut Cochin, INSERM U1016, UMR 8104, Paris, France; Department of Dermatology, Hôpital Cochin, AP-HP.Centre-Université Paris Cité, Paris, France; University Paris Cité, Faculté de Médecine Paris Centre Santé, Paris.
| | - Bénédicte Oulès
- Cutaneous Biology Lab, Institut Cochin, INSERM U1016, UMR 8104, Paris, France; Department of Dermatology, Hôpital Cochin, AP-HP.Centre-Université Paris Cité, Paris, France; University Paris Cité, Faculté de Médecine Paris Centre Santé, Paris
| |
Collapse
|
8
|
Shangaris P, El Hoss S. Fetal microchimerism and beyond: a new player in regenerative medicine. Haematologica 2023; 108:1731-1733. [PMID: 36700405 PMCID: PMC10316241 DOI: 10.3324/haematol.2022.282244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 01/10/2023] [Indexed: 01/27/2023] Open
Affiliation(s)
- Panicos Shangaris
- Women and Children's Health, School of Life Course and Population Sciences, Kings College London, London, United Kingdom; Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London.
| | - Sara El Hoss
- Red Cell Hematology Lab, Comprehensive Cancer Center, School of Cancer and Pharmaceutical Sciences, King's College London
| |
Collapse
|
9
|
du Halgouet A, Darbois A, Alkobtawi M, Mestdagh M, Alphonse A, Premel V, Yvorra T, Colombeau L, Rodriguez R, Zaiss D, El Morr Y, Bugaut H, Legoux F, Perrin L, Aractingi S, Golub R, Lantz O, Salou M. Role of MR1-driven signals and amphiregulin on the recruitment and repair function of MAIT cells during skin wound healing. Immunity 2023; 56:78-92.e6. [PMID: 36630919 PMCID: PMC9839364 DOI: 10.1016/j.immuni.2022.12.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 09/02/2022] [Accepted: 12/06/2022] [Indexed: 01/11/2023]
Abstract
Tissue repair processes maintain proper organ function following mechanical or infection-related damage. In addition to antibacterial properties, mucosal associated invariant T (MAIT) cells express a tissue repair transcriptomic program and promote skin wound healing when expanded. Herein, we use a human-like mouse model of full-thickness skin excision to assess the underlying mechanisms of MAIT cell tissue repair function. Single-cell RNA sequencing analysis suggested that skin MAIT cells already express a repair program at steady state. Following skin excision, MAIT cells promoted keratinocyte proliferation, thereby accelerating healing. Using skin grafts, parabiosis, and adoptive transfer experiments, we show that MAIT cells migrated into the wound in a T cell receptor (TCR)-independent but CXCR6 chemokine receptor-dependent manner. Amphiregulin secreted by MAIT cells following excision promoted wound healing. Expression of the repair function was probably independent of sustained TCR stimulation. Overall, our study provides mechanistic insights into MAIT cell wound healing function in the skin.
Collapse
Affiliation(s)
| | - Aurélie Darbois
- INSERM U932, PSL University, Institut Curie, 75005 Paris, France
| | - Mansour Alkobtawi
- Cutaneous Biology, Institut Cochin, Inserm 1016, and Université de Paris Cité, 75014 Paris, France
| | - Martin Mestdagh
- INSERM U932, PSL University, Institut Curie, 75005 Paris, France
| | - Aurélia Alphonse
- INSERM U932, PSL University, Institut Curie, 75005 Paris, France
| | - Virginie Premel
- INSERM U932, PSL University, Institut Curie, 75005 Paris, France
| | - Thomas Yvorra
- CNRS UMR 3666, INSERM U1143, Chemical Biology of Cancer Laboratory, PSL University, Institut Curie, 75005 Paris, France
| | - Ludovic Colombeau
- CNRS UMR 3666, INSERM U1143, Chemical Biology of Cancer Laboratory, PSL University, Institut Curie, 75005 Paris, France
| | - Raphaël Rodriguez
- CNRS UMR 3666, INSERM U1143, Chemical Biology of Cancer Laboratory, PSL University, Institut Curie, 75005 Paris, France
| | - Dietmar Zaiss
- Department of Immune Medicine, University of Regensburg, Regensburg, Germany,Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany,Institute of Pathology, University Regensburg, Regensburg, Germany,Leibniz Institute for Immunotherapy (LIT), Regensburg, Germany
| | - Yara El Morr
- INSERM U932, PSL University, Institut Curie, 75005 Paris, France
| | - Hélène Bugaut
- INSERM U932, PSL University, Institut Curie, 75005 Paris, France
| | - François Legoux
- INSERM U932, PSL University, Institut Curie, 75005 Paris, France
| | - Laetitia Perrin
- INSERM U932, PSL University, Institut Curie, 75005 Paris, France
| | - Selim Aractingi
- Cutaneous Biology, Institut Cochin, Inserm 1016, and Université de Paris Cité, 75014 Paris, France
| | - Rachel Golub
- Institut Pasteur, Université Paris Cité, INSERM U1223, 75015 Paris, France
| | - Olivier Lantz
- INSERM U932, PSL University, Institut Curie, 75005 Paris, France; Laboratoire d'Immunologie Clinique, Institut Curie, 75005 Paris, France; Centre d'investigation Clinique en Biothérapie Gustave-Roussy Institut Curie (CIC-BT1428), Institut Curie, 75005 Paris, France.
| | - Marion Salou
- INSERM U932, PSL University, Institut Curie, 75005 Paris, France.
| |
Collapse
|
10
|
Ribeiro RSDA, Demarque KC, Figueiredo Júnior I, Ferreira IMDESR, Valeriano JDP, Verícimo MA. Do Fetal Microchimeric Cells Influence Experimental Autoimmune Myocarditis? Fetal Pediatr Pathol 2022; 41:781-793. [PMID: 34678109 DOI: 10.1080/15513815.2021.1994067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Objective: We investigated the presence and influence of fetal microchimerism in the cardiac tissue of mated female mice submitted to experimental autoimmune myocarditis. Materials and methods: Nulliparous BALB/c females and BALB/c females mated with either BALB/c males (syngeneic mating) or C57BL/6 males (allogeneic mating) were immunized with cardiac myosin peptide MyHC-α614-629 or kept as non-immunized controls. Immunization occurred 6-8 weeks after delivery and mice were assessed after 21 days. Results: Immunized mice of allogeneic mating had a lower production of anti-MyHC-α614-629 antibodies compared to immunized nulliparous mice. Immunized nulliparous females had an intense mononuclear inflammatory infiltrate in cardiac tissue, associated with fibroplasia, while mated females had a lower inflammatory reaction. An increase in the frequency of microchimeric fetal cells was observed in mice submitted to allogeneic mating following immunization. Conclusion: Allogeneic cells of fetal origin could contribute to mitigating the inflammatory response in experimental myocarditis.
Collapse
Affiliation(s)
- Roberto Stefan de Almeida Ribeiro
- Department of Immunobiology, Institute of Biology, Federal Fluminense University, Niterói, Brazil.,Graduate Program in Pathology, Federal Fluminense University, Niterói, Brazil
| | | | - Israel Figueiredo Júnior
- Maternal and Child Department, Antônio Pedro University Hospital, Federal Fluminense University, Niterói, Brazil
| | | | - Jessica do Prado Valeriano
- Department of Immunobiology, Institute of Biology, Federal Fluminense University, Niterói, Brazil.,Graduate Program in Pathology, Federal Fluminense University, Niterói, Brazil
| | - Maurício Afonso Verícimo
- Department of Immunobiology, Institute of Biology, Federal Fluminense University, Niterói, Brazil.,Graduate Program in Pathology, Federal Fluminense University, Niterói, Brazil
| |
Collapse
|
11
|
Shi XQ, Chen G, Tan JQ, Li Z, Chen SM, He JH, Zhang L, Xu HX. Total alkaloid fraction of Leonurus japonicus Houtt. Promotes angiogenesis and wound healing through SRC/MEK/ERK signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2022; 295:115396. [PMID: 35598796 DOI: 10.1016/j.jep.2022.115396] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/12/2022] [Accepted: 05/16/2022] [Indexed: 06/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Leonurus japonicus Houtt., also known as motherwort, is a traditional Chinese medicine that was first identified in Shennong Bencao Jing, the first and essential pharmacy monograph in China. L. japonicus has been regarded as a good gynecological medicine since ancient times. It has been widely used in clinical settings for treatment of gynecological diseases and postnatal rehabilitation with good efficacy and low adverse effects. AIM OF THE STUDY The main purpose of this study was to determine the angiogenic and wound healing effects of total alkaloid fraction from L. japonicus Houtt. (TALH) in vivo and in vitro. In addition, the main bioactive components of total alkaloids were to be identified and analyzed in this study. MATERIALS AND METHODS First, the UHPLC/Q-TOF-MS method was used to identify and quantify the major components in the TALH extract. The wound healing activity was evaluated in vivo using a rat full-thickness cutaneous wound model. Histological study of wound healing in rat model was performed via immunohistochemistry and immunofluorescence. Cell proliferation was determined by MTT assay. Wound healing and transwell assays were used for detection of cell migration. The effect on tube formation was determined by tube formation assay in HUVECs. Western blot and RT-PCR were used to detect the expressions of relative proteins and genes respectively. Knock-down of SRC by siRNA was done to verify the crucial role of SRC in promotion of angiogenesis induced by TALH. RESULTS Seven characteristic peaks were recognized in the UHPLC/Q-TOF-MS spectrum, while four of the main components were quantified. The wound model in rats showed that treatment of TALH promoted wound healing by stimulating cellular proliferation and collagen deposition. In vitro experiments showed that co-treatment of TALH and VEGF increased cell proliferation, migration and tube formation in HUVECs. Mechanistic studies suggested that the co-treatment increased gene expressions of SRC, MEK1/2 and ERK1/2, as well as the phosphorylation levels of these proteins. Furthermore, the effect of co-treatment was attenuated after SRC knockdown, suggesting that SRC plays an important role in angiogenesis and wound healing induced by TALH and VEGF co-treatment. CONCLUSION Our results showed that TALH was one of the main active components of L. japonicus that promoted angiogenesis and wound healing by regulating the SRC/MEK/ERK pathway. Our study provided scientific basis for better clinical application of L. japonicas.
Collapse
Affiliation(s)
- Xiao-Qin Shi
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, PR China; Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, 201203, PR China
| | - Gan Chen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, PR China; Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, 201203, PR China
| | - Jia-Qi Tan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, PR China; Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, 201203, PR China
| | - Zhuo Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, PR China; Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, 201203, PR China
| | - Si-Min Chen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, PR China; Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, 201203, PR China
| | - Jia-Hui He
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, PR China; Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, 201203, PR China
| | - Li Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, PR China; Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai, 201203, PR China.
| | - Hong-Xi Xu
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
12
|
Xie Y, Yang H, Yang C, He L, Zhang X, Peng L, Zhu H, Gao L. Role and Mechanisms of Tumor-Associated Macrophages in Hematological Malignancies. Front Oncol 2022; 12:933666. [PMID: 35875135 PMCID: PMC9301190 DOI: 10.3389/fonc.2022.933666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 06/15/2022] [Indexed: 11/13/2022] Open
Abstract
Mounting evidence has revealed that many nontumor cells in the tumor microenvironment, such as fibroblasts, endothelial cells, mesenchymal stem cells, and leukocytes, are strongly involved in tumor progression. In hematological malignancies, tumor-associated macrophages (TAMs) are considered to be an important component that promotes tumor growth and can be polarized into different phenotypes with protumor or antitumor roles. This Review emphasizes research related to the role and mechanisms of TAMs in hematological malignancies. TAMs lead to poor prognosis by influencing tumor progression at the molecular level, including nurturing cancer stem cells and laying the foundation for metastasis. Although detailed molecular mechanisms have not been clarified, TAMs may be a new therapeutic target in hematological disease treatment.
Collapse
|
13
|
Sedov E, McCarthy J, Koren E, Fuchs Y. Fetomaternal microchimerism in tissue repair and tumor development. Dev Cell 2022; 57:1442-1452. [PMID: 35700729 DOI: 10.1016/j.devcel.2022.05.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 05/11/2022] [Accepted: 05/20/2022] [Indexed: 11/03/2022]
Abstract
In various placental mammals, the bidirectional exchange of cells during pregnancy can lead to the acquisition of genetically unique cells that can persist in both mother and child for decades. Over the years, it has become increasingly clear that this phenomenon, termed fetomaternal microchimerism may play key roles in a number of biological processes. In this perspective, we explore the concept of fetomaternal microchimerism and outline how fetal microchimeric cells are detected and immunologically tolerated within the maternal setting. Moreover, we discuss undertakings in the field that hint at the significant plasticity of fetal microchimeric cells and their potential roles in promoting maternal wound healing. Finally, we explore the multifaceted roles of fetal microchimeric cells in cancer development and progression. A deeper understanding of fetomaternal chimerism in healthy and diseased states will be key toward developing more efficient anti-cancer treatments and regenerative therapies.
Collapse
Affiliation(s)
- Egor Sedov
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion - Israel Institute of Technology, Haifa 3200003, Israel; Lorry Lokey Interdisciplinary Center for Life Sciences and Engineering, Technion Israel Institute of Technology, Haifa 3200003, Israel; Technion Integrated Cancer Center, Technion Israel Institute of Technology, Haifa 3200003, Israel
| | - Jordan McCarthy
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion - Israel Institute of Technology, Haifa 3200003, Israel; Lorry Lokey Interdisciplinary Center for Life Sciences and Engineering, Technion Israel Institute of Technology, Haifa 3200003, Israel; Technion Integrated Cancer Center, Technion Israel Institute of Technology, Haifa 3200003, Israel
| | - Elle Koren
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion - Israel Institute of Technology, Haifa 3200003, Israel; Lorry Lokey Interdisciplinary Center for Life Sciences and Engineering, Technion Israel Institute of Technology, Haifa 3200003, Israel; Technion Integrated Cancer Center, Technion Israel Institute of Technology, Haifa 3200003, Israel
| | - Yaron Fuchs
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion - Israel Institute of Technology, Haifa 3200003, Israel; Lorry Lokey Interdisciplinary Center for Life Sciences and Engineering, Technion Israel Institute of Technology, Haifa 3200003, Israel; Technion Integrated Cancer Center, Technion Israel Institute of Technology, Haifa 3200003, Israel.
| |
Collapse
|
14
|
IL-10 Dysregulation Underlies Chemokine Insufficiency, Delayed Macrophage Response, and Impaired Healing in Diabetic Wounds. J Invest Dermatol 2022; 142:692-704.e14. [PMID: 34517005 PMCID: PMC8860852 DOI: 10.1016/j.jid.2021.08.428] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 08/06/2021] [Accepted: 08/10/2021] [Indexed: 02/08/2023]
Abstract
Persistent inflammation is a major contributor to healing impairment in diabetic chronic wounds. Paradoxically, diabetic wound environment during the acute phase of healing is completely different because it exhibits a reduced macrophage response owing to inadequate expression of CCL2 proinflammatory cytokine. What causes a reduction in CCL2 expression in diabetic wounds early after injury remains unknown. In this study, we report that in contrast to prolonged exposure to high glucose, which makes monocytes proinflammatory, short-term exposure to high glucose causes a rapid monocyte reprogramming, manifested by increased expression and secretion of IL-10, which in an autocrine/paracrine fashion reduces glucose uptake and transforms monocytes into an anti-inflammatory phenotype by dampening signaling through toll-like receptors. We show that IL-10 expression is significantly increased in diabetic wounds during the acute phase of healing, causing significant reductions in toll-like receptor signaling and proinflammatory cytokine production, delaying macrophage and leukocyte responses, and underlying healing impairment in diabetic wounds. Importantly, blocking IL-10 signaling during the acute phase of healing improves toll-like receptor signaling, increases proinflammatory cytokine production, enhances macrophage and leukocyte responses, and stimulates healing in diabetic wounds. We posit that anti-IL-10 strategies have therapeutic potential if added topically after surgical debridement, which resets chronic wounds into acute fresh wounds.
Collapse
|
15
|
Wei CY, Li MQ, Zhu XY, Li DJ. Immune status of decidual macrophages is dependent on the CCL2/CCR2/JAK2 pathway during early pregnancy. Am J Reprod Immunol 2021; 86:e13480. [PMID: 34191381 DOI: 10.1111/aji.13480] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 06/22/2021] [Accepted: 06/28/2021] [Indexed: 12/13/2022] Open
Abstract
PROBLEM Decidual macrophages (dMφ ) play an important role in the formation of maternal-fetal immune tolerance. However, factors that influence the immune status of dMφ and the related potential mechanisms have not been elucidated to date. METHOD OF STUDY The gene transcription in dMφ , decidual stromal cells (DSCs), extravillous trophoblasts (EVTs), and peripheral monocytes (pMo) from human samples were measured using real-time polymerase chain reaction (PCR). Monocyte-DSC co-culture was established to explore whether DSCs influenced dMφ polarization via C-C motif ligand 2 (CCL2)-C-C chemokine receptor (CCR2) binding using flow cytometry. In vivo, changes in dMφ percentage and M1 and M2 marker expression after treatment with CCR2 or Janus kinase 2 (JAK2) inhibitor were detected with flow cytometry. Embryo resorption percentages in the above groups were also analyzed. RESULTS We found that dMφ were an M1/M2 mixed status at the maternal-fetal interface during early pregnancy. CCL2 influenced the immune status of dMφ in an autocrine and paracrine manner. As a downstream regulator of CCR2 and triggers the Stat3 pathway, JAK2 was found to be essential for dMφ homeostasis in vivo. JAK2 inhibitor decreased the dMφ proportion and attenuated Ki67, CD36, CD86, CD206, TNF, and IL-10 expression in dMφ at E8.5 d. Moreover, CCR2-JAK2 pathway inhibition decreased the width of the placental labyrinth layer, further influencing the pregnancy outcome. CONCLUSION The M1/M2 mixed immune status of dMφ was regulated by DSCs via CCR2, and the CCL2/CCR2/JAK2 pathway was essential for the immune status of dMφ and the outcome of early pregnancy.
Collapse
Affiliation(s)
- Chun-Yan Wei
- Laboratory for Reproductive Immunology, Hospital and Institute of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai, China
| | - Ming-Qing Li
- Laboratory for Reproductive Immunology, Hospital and Institute of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai, China
| | - Xiao-Yong Zhu
- Laboratory for Reproductive Immunology, Hospital and Institute of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Da-Jin Li
- Laboratory for Reproductive Immunology, Hospital and Institute of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| |
Collapse
|
16
|
Rosner M, Kolbe T, Voronin V, Hengstschläger M. Amniotic fluid stem cells and the cell source repertoire for non-invasive prenatal testing. Stem Cell Rev Rep 2021; 18:1366-1371. [PMID: 34383184 PMCID: PMC9033748 DOI: 10.1007/s12015-021-10228-5] [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] [Accepted: 07/26/2021] [Indexed: 12/04/2022]
Abstract
Cell-free fetal DNA (cffDNA)-based non-invasive prenatal testing (NIPT) is considered to be a very promising screening tool for pregnant women with an increased risk of fetal aneuploidy. Already millions of women worldwide underwent NIPT. However, due to the observed false-positive and false-negative results, this screening approach does not fulfil the criteria of a diagnostic test. Accordingly, positive results still require risk-carrying invasive prenatal testing, such as amniocentesis or chorionic villus sampling (CVS), for confirmation. Such hurdles need to be overcome before NIPT could become a diagnostic approach widely used in the general population. Here we discuss new evidence that besides the placenta amniotic fluid stem cells (AFSCs) could also represent an origin of cffDNA in the mother’s blood. A comprehensive picture of the involved cell source repertoire could pave the way to more reliable interpretations of NIPT results and ameliorate counselling of advice-seeking patients.
Collapse
Affiliation(s)
- Margit Rosner
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Währinger Strasse 10, 1090, Vienna, Austria
| | - Thomas Kolbe
- Biomodels Austria, University of Veterinary Medicine Vienna, Vienna, Austria.,Department IFA Tulln, University of Natural Resources and Life Sciences, Tulln, Austria
| | - Viktor Voronin
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Währinger Strasse 10, 1090, Vienna, Austria
| | - Markus Hengstschläger
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Währinger Strasse 10, 1090, Vienna, Austria.
| |
Collapse
|
17
|
Rosner M, Hengstschläger M. Three-dimensional migration of human amniotic fluid stem cells involves mesenchymal and amoeboid modes and is regulated by mTORC1. STEM CELLS (DAYTON, OHIO) 2021; 39:1718-1732. [PMID: 34331786 PMCID: PMC9291078 DOI: 10.1002/stem.3441] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 07/14/2021] [Indexed: 11/29/2022]
Abstract
Three‐dimensional (3D) cell migration is an integral part of many physiologic processes. Although being well studied in the context of adult tissue homeostasis and cancer development, remarkably little is known about the invasive behavior of human stem cells. Using two different kinds of invasion assays, this study aimed at investigating and characterizing the 3D migratory capacity of human amniotic fluid stem cells (hAFSCs), a well‐established fetal stem cell type. Eight hAFSC lines were found to harbor pronounced potential to penetrate basement membrane (BM)‐like matrices. Morphological examination and inhibitor approaches revealed that 3D migration of hAFSCs involves both the matrix metalloprotease‐dependent mesenchymal, elongated mode and the Rho‐associated protein kinase‐dependent amoeboid, round mode. Moreover, hAFSCs could be shown to harbor transendothelial migration capacity and to exhibit a motility‐associated marker expression pattern. Finally, the potential to cross extracellular matrix was found to be induced by mTORC1‐activating growth factors and reduced by blocking mTORC1 activity. Taken together, this report provides the first demonstration that human stem cells exhibit mTORC1‐dependent invasive capacity and can concurrently make use of mesenchymal and amoeboid 3D cell migration modes, which represents an important step toward the full biological characterization of fetal human stem cells with relevance to both developmental research and stem cell‐based therapy.
Collapse
Affiliation(s)
- Margit Rosner
- Institute of Medical Genetics, Center of Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
| | - Markus Hengstschläger
- Institute of Medical Genetics, Center of Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
18
|
Rosner M, Kolbe T, Hengstschläger M. Fetomaternal microchimerism and genetic diagnosis: On the origins of fetal cells and cell-free fetal DNA in the pregnant woman. MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2021; 788:108399. [PMID: 34893150 DOI: 10.1016/j.mrrev.2021.108399] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 11/11/2021] [Accepted: 11/14/2021] [Indexed: 06/14/2023]
Abstract
During pregnancy several types of fetal cells and fetal stem cells, including pregnancy-associated progenitor cells (PAPCs), traffic into the maternal circulation. Whereas they also migrate to various maternal organs and adopt the phenotype of the target tissues to contribute to regenerative processes, fetal cells also play a role in the pathogenesis of maternal diseases. In addition, cell-free fetal DNA (cffDNA) is detectable in the plasma of pregnant women. Together they constitute the well-known phenomenon of fetomaternal microchimerism, which inspired the concept of non-invasive prenatal testing (NIPT) using maternal blood. An in-depth knowledge concerning the origins of these fetal cells and cffDNA allows a more comprehensive understanding of the biological relevance of fetomaternal microchimerism and has implications for the ongoing expansion of resultant clinical applications.
Collapse
Affiliation(s)
- Margit Rosner
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
| | - Thomas Kolbe
- Biomodels Austria, University of Veterinary Medicine Vienna, Vienna, Austria; Department IFA Tulln, University of Natural Resources and Life Sciences, Tulln, Austria
| | - Markus Hengstschläger
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
19
|
Borkar R, Wang X, Zheng D, Miao Z, Zhang Z, Li E, Wu Y, Xu RH. Human ESC-derived MSCs enhance fat engraftment by promoting adipocyte reaggregation, secreting CCL2 and mobilizing macrophages. Biomaterials 2021; 272:120756. [PMID: 33798959 DOI: 10.1016/j.biomaterials.2021.120756] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 02/10/2021] [Accepted: 02/25/2021] [Indexed: 12/15/2022]
Abstract
Mesenchymal stem cells (MSCs) derived from somatic tissues have been used to promote lipotransfer, a common practice in cosmetic surgery. However, the effect of lipotransfer varies, and the mechanism of action remains vague. To address these questions, we differentiated human embryonic stem cells, a stable and unlimited source, into MSCs (EMSCs). Then we subcutaneously transplanted human fat aspirates together with EMSCs or PBS as a control into the back of nude mice. Within 24 h of transplantation, EMSCs promoted aggregation and encapsulation of injected fat tissues. Afterward, all grafts gradually shrank. However, EMSC-containing grafts were larger, heavier and had fewer dark areas on the surface than the control grafts. Histologically, more live adipocytes, vascular cells, and macrophages and less fibrosis were observed in EMSC-containing grafts than in the controls. Some EMSCs differentiated into vascular cells and adipocytes in the EMSC-containing grafts. RNA sequencing revealed that human RNA was shown to decline rapidly, while mouse RNA increased in the grafts; further, human genes related to extracellular matrix remodeling, adipogenesis, and chemokine (including CCL2) signaling were expressed at higher levels in the EMSC-containing grafts than they were in the controls. CCL2 knockout reduced macrophage migration towards EMSCs in vitro and early macrophage recruitment to the grafts and the pro-engraftment effect of EMSCs in vivo. Treating mice with a macrophage inhibitor abolished the EMSC effects and converted the grafts to heavy masses of cell debris. Together, these data demonstrate that EMSCs promote fat engraftment via enhanced tissue reconstitution and encapsulation of implanted tissues, which was followed by increased angiogenesis and adipocyte survival and reduced fibrosis, in which stimulated CCL2 signaling and mobilized macrophages play pivotal roles.
Collapse
Affiliation(s)
- Roma Borkar
- Center of Reproduction, Development & Aging, And Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Xiaoyan Wang
- Center of Reproduction, Development & Aging, And Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Dejin Zheng
- Center of Reproduction, Development & Aging, And Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Zhengqiang Miao
- Center of Reproduction, Development & Aging, And Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Zhenwu Zhang
- Center of Reproduction, Development & Aging, And Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Enqin Li
- Center of Reproduction, Development & Aging, And Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Yaojiong Wu
- The Shenzhen Key Laboratory of Health Sciences and Technology, International Graduate School at Shenzhen, Tsinghua University, Shenzhen, China
| | - Ren-He Xu
- Center of Reproduction, Development & Aging, And Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau, China.
| |
Collapse
|
20
|
Kornstädt L, Pierre S, Weigert A, Ebersberger S, Schäufele TJ, Kolbinger A, Schmid T, Cohnen J, Thomas D, Ferreirós N, Brüne B, Ebersberger I, Scholich K. Bacterial and Fungal Toll-Like Receptor Activation Elicits Type I IFN Responses in Mast Cells. Front Immunol 2021; 11:607048. [PMID: 33643293 PMCID: PMC7907501 DOI: 10.3389/fimmu.2020.607048] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 12/29/2020] [Indexed: 01/18/2023] Open
Abstract
Next to their role in IgE-mediated allergic diseases and in promoting inflammation, mast cells also have antiinflammatory functions. They release pro- as well as antiinflammatory mediators, depending on the biological setting. Here we aimed to better understand the role of mast cells during the resolution phase of a local inflammation induced with the Toll-like receptor (TLR)-2 agonist zymosan. Multiple sequential immunohistology combined with a statistical neighborhood analysis showed that mast cells are located in a predominantly antiinflammatory microenvironment during resolution of inflammation and that mast cell-deficiency causes decreased efferocytosis in the resolution phase. Accordingly, FACS analysis showed decreased phagocytosis of zymosan and neutrophils by macrophages in mast cell-deficient mice. mRNA sequencing using zymosan-induced bone marrow-derived mast cells (BMMC) revealed a strong type I interferon (IFN) response, which is known to enhance phagocytosis by macrophages. Both, zymosan and lipopolysaccharides (LPS) induced IFN-β synthesis in BMMCs in similar amounts as in bone marrow derived macrophages. IFN-β was expressed by mast cells in paws from naïve mice and during zymosan-induced inflammation. As described for macrophages the release of type I IFNs from mast cells depended on TLR internalization and endosome acidification. In conclusion, mast cells are able to produce several mediators including IFN-β, which are alone or in combination with each other able to regulate the phagocytotic activity of macrophages during resolution of inflammation.
Collapse
Affiliation(s)
- Lisa Kornstädt
- Institute of Clinical Pharmacology, University Hospital Goethe-University Frankfurt, Frankfurt, Germany
| | - Sandra Pierre
- Institute of Clinical Pharmacology, University Hospital Goethe-University Frankfurt, Frankfurt, Germany
| | - Andreas Weigert
- Faculty of Medicine, Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany
| | | | - Tim J. Schäufele
- Institute of Clinical Pharmacology, University Hospital Goethe-University Frankfurt, Frankfurt, Germany
| | - Anja Kolbinger
- Institute of Clinical Pharmacology, University Hospital Goethe-University Frankfurt, Frankfurt, Germany
| | - Tobias Schmid
- Faculty of Medicine, Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany
| | - Jennifer Cohnen
- Institute of Clinical Pharmacology, University Hospital Goethe-University Frankfurt, Frankfurt, Germany
| | - Dominique Thomas
- Institute of Clinical Pharmacology, University Hospital Goethe-University Frankfurt, Frankfurt, Germany
| | - Nerea Ferreirós
- Institute of Clinical Pharmacology, University Hospital Goethe-University Frankfurt, Frankfurt, Germany
| | - Bernhard Brüne
- Faculty of Medicine, Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany
- Project Group Translational Medicine and Pharmacology, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Frankfurt am Main, Germany
| | - Ingo Ebersberger
- Department for Applied Bioinformatics, Institute for Cell Biology and Neuroscience, Goethe-University Frankfurt, Frankfurt, Germany
- Senckenberg Biodiversity and Climate Research Centre (S-BIKF), Frankfurt am Main, Germany
- LOEWE Centre for Translational Biodiversity Genomics (TBG), Frankfurt am Main, Germany
| | - Klaus Scholich
- Institute of Clinical Pharmacology, University Hospital Goethe-University Frankfurt, Frankfurt, Germany
- Project Group Translational Medicine and Pharmacology, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD), Frankfurt am Main, Germany
| |
Collapse
|
21
|
Yang C, Luo Y, Lin H, Ge M, Shi J, Zhang X. Niobium Carbide MXene Augmented Medical Implant Elicits Bacterial Infection Elimination and Tissue Regeneration. ACS NANO 2021; 15:1086-1099. [PMID: 33372766 DOI: 10.1021/acsnano.0c08045] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Incurable bacterial infections, impenetrable microbial biofilm, and irreversible antibiotic resistance are among the most dangerous threats for humans. With few effective strategies available in antimicrobial and antibiofilm development, innovative methodologies inspired by the advances in other fields such as nanomedicine are becoming more and more attractive to realize innovative antibacterial agents. Herein, a 2D niobium carbide (Nb2C) MXene titanium plate (Nb2C@TP)-based clinical implant with practical multimodal anti-infection functions was developed. Such emerging modes are capable of destroying biofilms for direct bacteria elimination through down-regulating bacterial energy metabolism pathways, suppressing biofilm formation, and enhancing as-formed biofilm detachment via an activating accessory gene regulator. Another intriguing feature of this nanomedicine is the sensitization ability toward bacteria via photothermal transduction, which reduces the temperature necessary for bacteria eradication and mitigates possible normal tissue damage. Moreover, the Nb2C@TP medical implant is able to alleviate proinflammatory responses by scavenging excessive reactive oxygen species in infectious microenvironments, benefiting angiogenesis and tissue remodeling.
Collapse
Affiliation(s)
- Chuang Yang
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai 200233, P.R. China
| | - Yao Luo
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai 200233, P.R. China
| | - Han Lin
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics Chinese Academy of Sciences, Shanghai 200050, P.R. China
| | - Min Ge
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics Chinese Academy of Sciences, Shanghai 200050, P.R. China
| | - Jianlin Shi
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics Chinese Academy of Sciences, Shanghai 200050, P.R. China
| | - Xianlong Zhang
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai 200233, P.R. China
| |
Collapse
|
22
|
Yang C, Luo Y, Lin H, Ge M, Shi J, Zhang X. Niobium Carbide MXene Augmented Medical Implant Elicits Bacterial Infection Elimination and Tissue Regeneration. ACS NANO 2021; 15:1086-1099. [DOI: doi.org/10.1021/acsnano.0c08045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/08/2023]
Affiliation(s)
- Chuang Yang
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai 200233, P.R. China
| | - Yao Luo
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai 200233, P.R. China
| | - Han Lin
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics Chinese Academy of Sciences, Shanghai 200050, P.R. China
| | - Min Ge
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics Chinese Academy of Sciences, Shanghai 200050, P.R. China
| | - Jianlin Shi
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics Chinese Academy of Sciences, Shanghai 200050, P.R. China
| | - Xianlong Zhang
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai 200233, P.R. China
| |
Collapse
|
23
|
Bianchi DW, Khosrotehrani K, Way SS, MacKenzie TC, Bajema I, O'Donoghue K. Forever Connected: The Lifelong Biological Consequences of Fetomaternal and Maternofetal Microchimerism. Clin Chem 2020; 67:351-362. [PMID: 33417673 PMCID: PMC10072000 DOI: 10.1093/clinchem/hvaa304] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 10/28/2020] [Indexed: 01/07/2023]
Abstract
BACKGROUND Originally studied as a mechanism to understand eclampsia-related deaths during pregnancy, fetal cells in maternal blood have more recently garnered attention as a noninvasive source of fetal material for prenatal testing. In the 21st century, however, intact fetal cells have been largely supplanted by circulating cell-free placental DNA for aneuploidy screening. Instead, interest has pivoted to the ways in which fetal cells influence maternal biology. In parallel, an increasing appreciation of the consequences of maternal cells in the developing fetus has occurred. CONTENT In this review, we highlight the potential clinical applications and functional consequences of the bidirectional trafficking of intact cells between a pregnant woman and her fetus. Fetal cells play a potential role in the pathogenesis of maternal disease and tissue repair. Maternal cells play an essential role in educating the fetal immune system and as a factor in transplant acceptance. Naturally occurring maternal microchimerism is also being explored as a source of hematopoietic stem cells for transplant in fetal hematopoietic disorders. SUMMARY Future investigations in humans need to include complete pregnancy histories to understand maternal health and transplant success or failure. Animal models are useful to understand the mechanisms underlying fetal wound healing and/or repair associated with maternal injury and inflammation. The lifelong consequences of the exchange of cells between a mother and her child are profound and have many applications in development, health, and disease. This intricate exchange of genetically foreign cells creates a permanent connection that contributes to the survival of both individuals.
Collapse
Affiliation(s)
- Diana W Bianchi
- National Human Genome Research Institute and Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Kiarash Khosrotehrani
- Experimental Dermatology Group, The University of Queensland, UQ Diamantina Institute, Brisbane, Queensland, Australia
| | - Sing Sing Way
- Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Tippi C MacKenzie
- Center for Maternal-Fetal Precision Medicine and the Department of Surgery, University of California, San Francisco, CA, USA
| | - Ingeborg Bajema
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Keelin O'Donoghue
- Irish Centre for Maternal and Child Health (INFANT), University College Cork, Cork, Ireland
| |
Collapse
|
24
|
Zhou F, Liu F, Liu J, He YL, Zhou QM, Guo L, Peng C, Xiong L. Stachydrine promotes angiogenesis by regulating the VEGFR2/MEK/ERK and mitochondrial-mediated apoptosis signaling pathways in human umbilical vein endothelial cells. Biomed Pharmacother 2020; 131:110724. [DOI: 10.1016/j.biopha.2020.110724] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 08/28/2020] [Accepted: 09/02/2020] [Indexed: 12/14/2022] Open
|
25
|
Vadakke-Madathil S, Chaudhry HW. Chimerism as the basis for organ repair. Ann N Y Acad Sci 2020; 1487:12-20. [PMID: 32991755 DOI: 10.1111/nyas.14488] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 08/11/2020] [Accepted: 08/18/2020] [Indexed: 01/14/2023]
Abstract
Organ and tissue repair are complex processes involving signaling molecules, growth factors, and cell cycle regulators that act in concert to promote cell division and differentiation at sites of injury. In embryonic development, progenitor fetal cells are actively involved in reparative mechanisms and display a biphasic interaction with the mother; and there is constant trafficking of fetal cells into maternal circulation and vice versa. This phenomenon of fetal microchimerism may have significant impact considering the primitive, multilineage nature of these cells. In published work, we have reported that fetal-derived placental cells expressing the homeodomain protein CDX2 retain all "stem" functional proteins of embryonic stem cells yet are endowed with additional functions in areas of growth, survival, homing, and immune modulation. These cells exhibit multipotency in vitro and in vivo, giving rise to spontaneously beating cardiomyocytes and vascular cells. In mouse models, CDX2 cells from female placentas can be administered intravenously to male mice subjected to myocardial infarction with subsequent homing of the CDX2 cells to infarcted areas and evidence of cellular regeneration with enhanced cardiac function. Elucidating the role of microchimeric fetal-derived placental cells may have broader scientific potential, as one can envision allogeneic cell therapy strategies targeted at tissue regeneration for a variety of organ systems.
Collapse
Affiliation(s)
| | - Hina W Chaudhry
- Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
26
|
Blanc RS, Kallenbach JG, Bachman JF, Mitchell A, Paris ND, Chakkalakal JV. Inhibition of inflammatory CCR2 signaling promotes aged muscle regeneration and strength recovery after injury. Nat Commun 2020; 11:4167. [PMID: 32820177 PMCID: PMC7441393 DOI: 10.1038/s41467-020-17620-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 07/06/2020] [Indexed: 02/07/2023] Open
Abstract
Muscle regeneration depends on a robust albeit transient inflammatory response. Persistent inflammation is a feature of age-related regenerative deficits, yet the underlying mechanisms are poorly understood. Here, we find inflammatory-related CC-chemokine-receptor 2 (Ccr2) expression in non-hematopoietic myogenic progenitors (MPs) during regeneration. After injury, the expression of Ccr2 in MPs corresponds to the levels of its ligands, the chemokines Ccl2, 7, and 8. We find stimulation of Ccr2-activity inhibits MP fusion and contribution to myofibers. This occurs in association with increases in MAPKp38δ/γ signaling, MyoD phosphorylation, and repression of the terminal myogenic commitment factor Myogenin. High levels of Ccr2-chemokines are a feature of regenerating aged muscle. Correspondingly, deletion of Ccr2 in MPs is necessary for proper fusion into regenerating aged muscle. Finally, opportune Ccr2 inhibition after injury enhances aged regeneration and functional recovery. These results demonstrate that inflammatory-induced activation of Ccr2 signaling in myogenic cells contributes to aged muscle regenerative decline. Chronic inflammation is a feature of age-related regenerative decline in skeletal muscles, but how it directly affects resident muscle stem cell fate and function is unclear. Here, the authors show that Ccr2 signaling in muscle stem cell derived progenitors represses terminal myogenic differentiation, and that targeting Ccr2 on aged myogenic progenitors rejuvenates aged skeletal muscle healing and function.
Collapse
Affiliation(s)
- Roméo S Blanc
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA.,Wilmot Cancer Institute, Stem Cell and Regenerative Medicine Institute, and The Rochester Aging Research Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Jacob G Kallenbach
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA.,Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, USA
| | - John F Bachman
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA.,Department of Pathology and Laboratory Medicine, Cell Biology of Disease Graduate Program, University of Rochester Medical Center, Rochester, NY, USA
| | - Amanda Mitchell
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Nicole D Paris
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA.,Wilmot Cancer Institute, Stem Cell and Regenerative Medicine Institute, and The Rochester Aging Research Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Joe V Chakkalakal
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA. .,Wilmot Cancer Institute, Stem Cell and Regenerative Medicine Institute, and The Rochester Aging Research Center, University of Rochester Medical Center, Rochester, NY, USA. .,Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
27
|
Beksac MS, Fadiloglu E, Cakar AN, Gurbuz RH, Atilla P, Onbasilar I, Beksac K, Katlan DC, Mumusoglu S, Calis P, Beksac M. Fetal Cell Microchimerism; Normal and Immunocompromised Gestations in Mice. Fetal Pediatr Pathol 2020; 39:277-287. [PMID: 31436120 DOI: 10.1080/15513815.2019.1651803] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Objective: To compare fetal cell microchimerism in normal and immunocompromised gestations. Materials and methods: The study consists of two groups of mature female mice. In the control group and the immunocompromised study group, 5 mg of saline and cyclosporine were injected intraperitoneally, respectively. In the second step, all female mice were mated with "Actine-Luc (+) green fluorescent protein (GFP)" transgenic male mice. Immunohistochemical studies (ALPL-antiluciferase, cytokeratin-antiluciferase, and CD 105-antiluciferase) were carried out on maternal liver, skin, and lung tissues at 6-7th and 14-15th gestational days, and postpartum 3-4th, 12th, and 18-24 months. Results: GFP (+) cells were detected in maternal liver and skin but not in lung tissue. Liver was the most affected tissue. GFP was found to be more intense in the immunocompromised group. Conclusion: Fetal microchimerism was demonstrated in maternal liver and skin and found to be more intensive in the immunocompromised group.
Collapse
Affiliation(s)
- M Sinan Beksac
- Division of Perinatal Medicine, Department of Obstetrics and Gynecology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Erdem Fadiloglu
- Division of Perinatal Medicine, Department of Obstetrics and Gynecology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Ayse Nur Cakar
- Department of Histology and Embryology, TOBB University Faculty of Medicine, Ankara, Turkey
| | - Rumeysa Hekimoglu Gurbuz
- Department of Histology and Embryology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Pergin Atilla
- Department of Histology and Embryology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Ilyas Onbasilar
- Animal Technology Center, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Kemal Beksac
- Department of General Surgery, Dr. Abdurrahman Yurtaslan Oncology Education and Research Hospital, Ankara, Turkey
| | - Doruk Cevdi Katlan
- Division of Perinatal Medicine, Department of Obstetrics and Gynecology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Sezcan Mumusoglu
- Division of Perinatal Medicine, Department of Obstetrics and Gynecology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Pinar Calis
- Division of Perinatal Medicine, Department of Obstetrics and Gynecology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Meral Beksac
- Department of Haematology, Faculty of Medicine, Ankara University, Ankara, Turkey
| |
Collapse
|
28
|
PDGFB-expressing mesenchymal stem cells improve human hematopoietic stem cell engraftment in immunodeficient mice. Bone Marrow Transplant 2019; 55:1029-1040. [PMID: 31804621 PMCID: PMC7269905 DOI: 10.1038/s41409-019-0766-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 11/13/2019] [Accepted: 11/21/2019] [Indexed: 12/11/2022]
Abstract
The bone marrow (BM) niche regulates multiple hematopoietic stem cell (HSC) processes. Clinical treatment for hematological malignancies by HSC transplantation often requires preconditioning via total body irradiation, which severely and irreversibly impairs the BM niche and HSC regeneration. Novel strategies are needed to enhance HSC regeneration in irradiated BM. We compared the effects of EGF, FGF2, and PDGFB on HSC regeneration using human mesenchymal stem cells (MSCs) that were transduced with these factors via lentiviral vectors. Among the above niche factors tested, MSCs transduced with PDGFB (PDGFB-MSCs) most significantly improved human HSC engraftment in immunodeficient mice. PDGFB-MSC-treated BM enhanced transplanted human HSC self-renewal in secondary transplantations more efficiently than GFP-transduced MSCs (GFP-MSCs). Gene set enrichment analysis showed increased antiapoptotic signaling in PDGFB-MSCs compared with GFP-MSCs. PDGFB-MSCs exhibited enhanced survival and expansion after transplantation, resulting in an enlarged humanized niche cell pool that provide a better humanized microenvironment to facilitate superior engraftment and proliferation of human hematopoietic cells. Our studies demonstrate the efficacy of PDGFB-MSCs in supporting human HSC engraftment.
Collapse
|
29
|
Do DC, Mu J, Ke X, Sachdeva K, Qin Z, Wan M, Ishmael FT, Gao P. miR-511-3p protects against cockroach allergen-induced lung inflammation by antagonizing CCL2. JCI Insight 2019; 4:126832. [PMID: 31536479 DOI: 10.1172/jci.insight.126832] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 09/12/2019] [Indexed: 02/06/2023] Open
Abstract
miR-511-3p, encoded by CD206/Mrc1, was demonstrated to reduce allergic inflammation and promote alternative (M2) macrophage polarization. Here, we sought to elucidate the fundamental mechanism by which miR-511-3p attenuates allergic inflammation and promotes macrophage polarization. Compared with WT mice, the allergen-challenged Mrc1-/- mice showed increased airway hyperresponsiveness (AHR) and inflammation. However, this increased AHR and inflammation were significantly attenuated when these mice were pretransduced with adeno-associated virus-miR-511-3p (AAV-miR-511-3p). Gene expression profiling of macrophages identified Ccl2 as one of the major genes that was highly expressed in M2 macrophages but antagonized by miR-511-3p. The interaction between miR-511-3p and Ccl2 was confirmed by in silico analysis and mRNA-miR pulldown assay. Further evidence for the inhibition of Ccl2 by miR-511-3p was given by reduced levels of Ccl2 in supernatants of miR-511-3p-transduced macrophages and in bronchoalveolar lavage fluids of AAV-miR-511-3p-infected Mrc1-/- mice. Mechanistically, we demonstrated that Ccl2 promotes M1 macrophage polarization by activating RhoA signaling through Ccr2. The interaction between Ccr2 and RhoA was also supported by coimmunoprecipitation assay. Importantly, inhibition of RhoA signaling suppressed cockroach allergen-induced AHR and lung inflammation. These findings suggest a potentially novel mechanism by which miR-511-3p regulates allergic inflammation and macrophage polarization by targeting Ccl2 and its downstream Ccr2/RhoA axis.
Collapse
Affiliation(s)
- Danh C Do
- Johns Hopkins Asthma and Allergy Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jie Mu
- Johns Hopkins Asthma and Allergy Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Anesthesiology, West China School of Medicine, Sichuan University, Chengdu, China
| | - Xia Ke
- Department of Otorhinolaryngology, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Karan Sachdeva
- Johns Hopkins Asthma and Allergy Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Zili Qin
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Mei Wan
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Faoud T Ishmael
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Peisong Gao
- Johns Hopkins Asthma and Allergy Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
30
|
CCL2 promotes macrophages-associated chemoresistance via MCPIP1 dual catalytic activities in multiple myeloma. Cell Death Dis 2019; 10:781. [PMID: 31611552 PMCID: PMC6791869 DOI: 10.1038/s41419-019-2012-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 08/27/2019] [Accepted: 09/19/2019] [Indexed: 02/07/2023]
Abstract
We previously showed that the chemokine CCL2 can recruit macrophages (Mφs) to the bone marrow (BM) in multiple myeloma (MM) and that myeloma-associated Mφs are important in drug resistance. Here, we explore the role of increased CCL2 expression in the BM microenvironment of MM and elucidate the underlying mechanism. Our results show that CCL2 expression is associated with the treatment status of MM patients. Mφs interact with MM cells and further upregulate their expression of CCL2. These increased level of CCL2 polarizes Mφs toward the M2-like phenotype and promotes Mφs to protect MM cells from drug-induced apoptosis. Mechanistically, CCL2 upregulated the expression of the immunosuppressive molecular MCP-1-induced protein (MCPIP1) in Mφs. MCPIP1 mediates Mφs’ polarization and protection via dual catalytic activities. Additionally, we found that CCL2 induces MCPIP1 expression via the JAK2-STAT3 signaling pathway. Taken together, our results indicate that increased CCL2 expression in MM patients’ BM polarizes Mφs toward the M2-like phenotype and promotes the protective effect of Mφs through MCPIP1, providing novel insight into the mechanism of Mφs-mediated drug resistance in MM.
Collapse
|
31
|
Ståhlberg A, El-Heliebi A, Sedlmayr P, Kroneis T. Unravelling the biological secrets of microchimerism by single-cell analysis. Brief Funct Genomics 2018; 17:255-264. [PMID: 29028900 PMCID: PMC6063264 DOI: 10.1093/bfgp/elx027] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The presence of microchimeric cells is known for >100 years and well documented since decades. Earlier, microchimeric cells were mainly used for cell-based non-invasive prenatal diagnostics during early pregnancy. Microchimeric cells are also present beyond delivery and are associated to various autoimmune diseases, tissue repair, cancer and immune tolerance. All these findings were based on low complexity studies and occasionally accompanied by artefacts not allowing the biological functions of microchimerism to be determined. However, with the recent developments in single-cell analysis, new means to identify and characterize microchimeric cells are available. Cell labelling techniques in combination with single-cell analysis provide a new toolbox to decipher the biology of microchimeric cells at molecular and cellular level. In this review, we discuss how recent developments in single-cell analysis can be applied to determine the role and function of microchimeric cells.
Collapse
Affiliation(s)
- Anders Ståhlberg
- Sahlgrenska Cancer Center, Department of Pathology and Genetics, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Medicinaregatan 1F, Gothenburg, Sweden
| | - Amin El-Heliebi
- Institute of Cell Biology, Histology & Embryology, Medical University of Graz, Harrachgasse 21, Graz, Austria
| | - Peter Sedlmayr
- Institute of Cell Biology, Histology & Embryology, Medical University of Graz, Harrachgasse 21, Graz, Austria
| | - Thomas Kroneis
- Sahlgrenska Cancer Center, Department of Pathology and Genetics, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Medicinaregatan 1F, Gothenburg, Sweden
- Institute of Cell Biology, Histology & Embryology, Medical University of Graz, Harrachgasse 21, Graz, Austria
| |
Collapse
|
32
|
Yoshimura T. The chemokine MCP-1 (CCL2) in the host interaction with cancer: a foe or ally? Cell Mol Immunol 2018; 15:335-345. [PMID: 29375123 DOI: 10.1038/cmi.2017.135] [Citation(s) in RCA: 191] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Revised: 10/13/2017] [Accepted: 10/18/2017] [Indexed: 12/13/2022] Open
Abstract
Macrophages are one of the most abundant leukocyte populations infiltrating tumor tissues and can exhibit both tumoricidal and tumor-promoting activities. In 1989, we reported the purification of monocyte chemoattractant protein-1 (MCP-1) from culture supernatants of mitogen-activated peripheral blood mononuclear cells and tumor cells. MCP-1 is a potent monocyte-attracting chemokine, identical to the previously described lymphocyte-derived chemotactic factor or tumor-derived chemotactic factor, and greatly contributes to the recruitment of blood monocytes into sites of inflammatory responses and tumors. Because in vitro-cultured tumor cells often produce significant amounts of MCP-1, tumor cells are considered to be the main source of MCP-1. However, various non-tumor cells in the tumor stroma also produce MCP-1 in response to stimuli. Studies performed in vitro and in vivo have provided evidence that MCP-1 production in tumors is a consequence of complex interactions between tumor cells and non-tumor cells and that both tumor cells and non-tumor cells contribute to the production of MCP-1. Although MCP-1 production was once considered to be a part of host defense against tumors, it is now believed to regulate the vicious cycle between tumor cells and macrophages that promotes the progression of tumors.
Collapse
Affiliation(s)
- Teizo Yoshimura
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 700-8558, Kita-ku, Okayama, Japan.
| |
Collapse
|
33
|
Cancer. CHIMERISM 2018. [DOI: 10.1007/978-3-319-89866-7_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
34
|
[What's new in dermatological research?]. Ann Dermatol Venereol 2017; 144 Suppl 4:IVS16-IVS22. [PMID: 29249248 DOI: 10.1016/s0151-9638(17)31061-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
This manuscript provides a selection of dermatological research manuscripts published from September 2016 to August 2017. It is not an exhaustive review but rather a selection of manuscripts susceptible to modify the dermatological practice or affording new pathophysiologic mechanisms and new therapeutic approaches. The following areas of interest are concerned: recognition of dermatological images by artificial intelligence, new concepts in atopic dermatitis, wound repair and hair growth cycle. New data concerning melanomagenesis, epidermolysis bullosa simplex and drug eruption are also highlighted.
Collapse
|
35
|
Lim W, Bae H, Bazer FW, Kim SM, Song G. C-C motif chemokine ligand 2 regulates lps-induced inflammation and ER stress to enhance proliferation of bovine endometrial epithelial cells. J Cell Physiol 2017; 233:3141-3151. [DOI: 10.1002/jcp.26151] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 08/11/2017] [Indexed: 12/13/2022]
Affiliation(s)
- Whasun Lim
- Department of Biomedical Sciences; Catholic Kwandong University; Gangneung Republic of Korea
| | - Hyocheol Bae
- Institute of Animal Molecular Biotechnology and Department of Biotechnology; College of Life Sciences and Biotechnology; Korea University; Seoul Republic of Korea
| | - Fuller W. Bazer
- Center for Animal Biotechnology and Genomics and Department of Animal Science; Texas A&M University; College Station Texas
| | - Sung-Man Kim
- Department of Medical Management; Catholic Kwandong University; Gangneung Republic of Korea
| | - Gwonhwa Song
- Institute of Animal Molecular Biotechnology and Department of Biotechnology; College of Life Sciences and Biotechnology; Korea University; Seoul Republic of Korea
| |
Collapse
|