1
|
Medina I, Wieland EB, Temmerman L, Otten JJT, Bermudez B, Bot I, Rademakers T, Wijnands E, Schurgers L, Mees B, van Berkel TJC, Goossens P, Biessen EAL. Colony stimulating factor 1 receptor (Csf1r) expressing cell ablation in mafia (macrophage-specific Fas-induced apoptosis) mice alters monocyte landscape and atherosclerotic lesion characteristics. Eur J Immunol 2024; 54:e2350943. [PMID: 39233527 DOI: 10.1002/eji.202350943] [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: 03/21/2024] [Revised: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 09/06/2024]
Abstract
Macrophage infiltration and accumulation in the atherosclerotic lesion are associated with plaque progression and instability. Depletion of macrophages from the lesion might provide valuable insights into plaque stabilization processes. Therefore, we assessed the effects of systemic and local macrophage depletion on atherogenesis. To deplete monocytes/macrophages we used atherosclerosis-susceptible Apoe- /- mice, bearing a MaFIA (macrophage-Fas-induced-apoptosis) suicide construct under control of the Csf1r (CD115) promotor, where selective apoptosis of Csf1r-expressing cells was induced in a controlled manner, by administration of a drug, AP20187. Systemic induction of apoptosis resulted in a decrease in lesion macrophages and smooth-muscle cells. Plaque size and necrotic core size remained unaffected. Two weeks after the systemic depletion of macrophages, we observed a replenishment of the myeloid compartment. Myelopoiesis was modulated resulting in an expansion of CSF1Rlo myeloid cells in the circulation and a shift from Ly6chi monocytes toward Ly6cint and Ly6clo populations in the spleen. Local apoptosis induction led to a decrease in plaque burden and macrophage content with marginal effects on the circulating myeloid cells. Local, but not systemic depletion of Csf1r+ myeloid cells resulted in decreased plaque burden. Systemic depletion led to CSF1Rlo-monocyte expansion in blood, possibly explaining the lack of effects on plaque development.
Collapse
Affiliation(s)
- Indira Medina
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
- Division of Biopharmaceutics, Leiden Academic Center for Drug Research, Leiden, the Netherlands
| | - Elias B Wieland
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Lieve Temmerman
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Jeroen J T Otten
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Beatriz Bermudez
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Ilze Bot
- Division of Biopharmaceutics, Leiden Academic Center for Drug Research, Leiden, the Netherlands
| | - Timo Rademakers
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, the Netherlands
| | - Erwin Wijnands
- Central Diagnostic Laboratory, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Leon Schurgers
- Department of Biochemistry, Maastricht University, Maastricht, the Netherlands
| | - Barend Mees
- Department of Vascular Surgery, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Theo J C van Berkel
- Division of Biopharmaceutics, Leiden Academic Center for Drug Research, Leiden, the Netherlands
| | - Pieter Goossens
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Erik A L Biessen
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Aachen, Germany
| |
Collapse
|
2
|
Gomez PT, Carver CM, Rodriguez SL, Wang L, Zhang X, Schafer MJ. Aging and senescent fates of oligodendrocyte precursor cells in the mouse brain. NPJ AGING 2024; 10:47. [PMID: 39438481 PMCID: PMC11496697 DOI: 10.1038/s41514-024-00176-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 09/29/2024] [Indexed: 10/25/2024]
Abstract
Age-related changes in oligodendrocyte precursor cells (OPCs) contribute to white matter dysfunction. In aged mice, we hypothesized that myelin-dense fimbria OPCs possess niche-specific properties, compared to hippocampal OPCs. Aged fimbria OPCs were fewer, larger, and localized to neighboring microglia. We identified age-increased p16/Cdkn2a-expressing OPCs enriched for senescence-related pathways and distinct senescence signatures between hippocampus and fimbria. Aged brain OPC populations differ in microenvironment properties and responses to senescence-directed intervention.
Collapse
Affiliation(s)
- Paul T Gomez
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Chase M Carver
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Sonia L Rodriguez
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Liguo Wang
- Division of Computational Biology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Xu Zhang
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Marissa J Schafer
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA.
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA.
- Department of Neurology, Mayo Clinic, Rochester, MN, USA.
- Department of Neuroscience, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
3
|
Brum ES, Fialho MFP, Souza Monteiro de Araújo D, Landini L, Marini M, Titiz M, Kuhn BL, Frizzo CP, Araújo PHS, Guimarães RM, Cunha TM, Silva CR, Trevisan G, Geppetti P, Nassini R, De Logu F, Oliveira SM. Schwann cell TRPA1 elicits reserpine-induced fibromyalgia pain in mice. Br J Pharmacol 2024; 181:3445-3461. [PMID: 38772415 DOI: 10.1111/bph.16413] [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: 11/23/2023] [Revised: 02/29/2024] [Accepted: 03/30/2024] [Indexed: 05/23/2024] Open
Abstract
BACKGROUND AND PURPOSE Fibromyalgia is a complex clinical disorder with an unknown aetiology, characterized by generalized pain and co-morbid symptoms such as anxiety and depression. An imbalance of oxidants and antioxidants is proposed to play a pivotal role in the pathogenesis of fibromyalgia symptoms. However, the precise mechanisms by which oxidative stress contributes to fibromyalgia-induced pain remain unclear. The transient receptor potential ankyrin 1 (TRPA1) channel, known as both a pain sensor and an oxidative stress sensor, has been implicated in various painful conditions. EXPERIMENTAL APPROACH The feed-forward mechanism that implicates reactive oxygen species (ROS) driven by TRPA1 was investigated in a reserpine-induced fibromyalgia model in C57BL/6J mice employing pharmacological interventions and genetic approaches. KEY RESULTS Reserpine-treated mice developed pain-like behaviours (mechanical/cold hypersensitivity) and early anxiety-depressive-like disorders, accompanied by increased levels of oxidative stress markers in the sciatic nerve tissues. These effects were not observed upon pharmacological blockade or global genetic deletion of the TRPA1 channel and macrophage depletion. Furthermore, we demonstrated that selective silencing of TRPA1 in Schwann cells reduced reserpine-induced neuroinflammation (NADPH oxidase 1-dependent ROS generation and macrophage increase in the sciatic nerve) and attenuated fibromyalgia-like behaviours. CONCLUSION AND IMPLICATIONS Activated Schwann cells expressing TRPA1 promote an intracellular pathway culminating in the release of ROS and recruitment of macrophages in the mouse sciatic nerve. These cellular and molecular events sustain mechanical and cold hypersensitivity in the reserpine-evoked fibromyalgia model. Targeting TRPA1 channels on Schwann cells could offer a novel therapeutic approach for managing fibromyalgia-related behaviours.
Collapse
Affiliation(s)
- Evelyne Silva Brum
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Centre of Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, Brazil
| | - Maria Fernanda Pessano Fialho
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Centre of Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, Brazil
| | | | - Lorenzo Landini
- Department of Health Sciences, Clinical Pharmacology Unit, University of Florence, Florence, Italy
| | - Matilde Marini
- Department of Health Sciences, Clinical Pharmacology Unit, University of Florence, Florence, Italy
| | - Mustafa Titiz
- Department of Health Sciences, Clinical Pharmacology Unit, University of Florence, Florence, Italy
| | - Bruna Luiza Kuhn
- Heterocycle Chemistry Nucleus (NUQUIMHE), Federal University of Santa Maria, Santa Maria, Brazil
| | - Clarissa Piccinin Frizzo
- Heterocycle Chemistry Nucleus (NUQUIMHE), Federal University of Santa Maria, Santa Maria, Brazil
| | | | - Rafaela Mano Guimarães
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Thiago Mattar Cunha
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Cássia Regina Silva
- Department of Genetic and Biochemistry, University of Uberlândia, Uberlândia, Brazil
| | - Gabriela Trevisan
- Graduate Program in Pharmacology, Federal University of Santa Maria, Santa Maria, Brazil
| | - Pierangelo Geppetti
- Department of Health Sciences, Clinical Pharmacology Unit, University of Florence, Florence, Italy
| | - Romina Nassini
- Department of Health Sciences, Clinical Pharmacology Unit, University of Florence, Florence, Italy
| | - Francesco De Logu
- Department of Health Sciences, Clinical Pharmacology Unit, University of Florence, Florence, Italy
| | - Sara Marchesan Oliveira
- Graduate Program in Biological Sciences: Toxicological Biochemistry, Centre of Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, Brazil
| |
Collapse
|
4
|
Geraghty T, Ishihara S, Obeidat AM, Adamczyk NS, Hunter RS, Li J, Wang L, Lee H, Ko FC, Malfait AM, Miller RE. Acute systemic macrophage depletion in osteoarthritic mice alleviates pain-related behaviors and does not affect joint damage. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.16.608301. [PMID: 39229102 PMCID: PMC11370380 DOI: 10.1101/2024.08.16.608301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Background Osteoarthritis (OA) is a painful degenerative joint disease and a leading source of years lived with disability globally due to inadequate treatment options. Neuroimmune interactions reportedly contribute to OA pain pathogenesis. Notably, in rodents, macrophages in the DRG are associated with onset of persistent OA pain. Our objective was to determine the effects of acute systemic macrophage depletion on pain-related behaviors and joint damage using surgical mouse models in both sexes. Methods We depleted CSF1R+ macrophages by treating male macrophage Fas-induced apoptosis (MaFIA) transgenic mice 8- or 16-weeks post destabilization of the medial meniscus (DMM) with AP20187 or vehicle control (10 mg/kg i.p., 1x/day for 5 days), or treating female MaFIA mice 12 weeks post partial meniscectomy (PMX) with AP20187 or vehicle control. We measured pain-related behaviors 1-3 days before and after depletion, and, 3-4 days after the last injection we examined joint histopathology and performed flow cytometry of the dorsal root ganglia (DRGs). In a separate cohort of male 8-week DMM mice or age-matched naïve vehicle controls, we conducted DRG bulk RNA-sequencing analyses after the 5-day vehicle or AP20187 treatment. Results Eight- and 16-weeks post DMM in male mice, AP20187-induced macrophage depletion resulted in attenuated mechanical allodynia and knee hyperalgesia. Female mice showed alleviation of mechanical allodynia, knee hyperalgesia, and weight bearing deficits after macrophage depletion at 12 weeks post PMX. Macrophage depletion did not affect the degree of cartilage degeneration, osteophyte width, or synovitis in either sex. Flow cytometry of the DRG revealed that macrophages and neutrophils were reduced after AP20187 treatment. In addition, in the DRG, only MHCII+ M1-like macrophages were significantly decreased, while CD163+MHCII- M2-like macrophages were not affected in both sexes. DRG bulk RNA-seq revealed that Cxcl10 and Il1b were upregulated with DMM surgery compared to naïve mice, and downregulated in DMM after acute macrophage depletion. Conclusions Acute systemic macrophage depletion reduced the levels of pro-inflammatory macrophages in the DRG and alleviated pain-related behaviors in established surgically induced OA in mice of both sexes, without affecting joint damage. Overall, these studies provide insight into immune cell regulation in the DRG during OA.
Collapse
Affiliation(s)
- Terese Geraghty
- Rush University Medical Center, Department of Internal Medicine, Division of Rheumatology, Chicago, IL USA
- Rush University Medical Center, Department of Anatomy & Cell Biology, Chicago, IL USA
| | - Shingo Ishihara
- Rush University Medical Center, Department of Internal Medicine, Division of Rheumatology, Chicago, IL USA
- Rush University Medical Center, Department of Anatomy & Cell Biology, Chicago, IL USA
| | - Alia M Obeidat
- Rush University Medical Center, Department of Internal Medicine, Division of Rheumatology, Chicago, IL USA
- Rush University Medical Center, Department of Anatomy & Cell Biology, Chicago, IL USA
| | - Natalie S Adamczyk
- Rush University Medical Center, Department of Internal Medicine, Division of Rheumatology, Chicago, IL USA
- Rush University Medical Center, Department of Anatomy & Cell Biology, Chicago, IL USA
| | - Rahel S Hunter
- Rush University Medical Center, Department of Internal Medicine, Division of Rheumatology, Chicago, IL USA
- Rush University Medical Center, Department of Anatomy & Cell Biology, Chicago, IL USA
| | - Jun Li
- Rush University Medical Center, Department of Internal Medicine, Division of Rheumatology, Chicago, IL USA
- Rush University Medical Center, Department of Anatomy & Cell Biology, Chicago, IL USA
| | - Lai Wang
- Rush University Medical Center, Department of Internal Medicine, Division of Rheumatology, Chicago, IL USA
- Rush University Medical Center, Department of Anatomy & Cell Biology, Chicago, IL USA
| | - Hoomin Lee
- Rush University Medical Center, Department of Internal Medicine, Division of Rheumatology, Chicago, IL USA
- Rush University Medical Center, Department of Anatomy & Cell Biology, Chicago, IL USA
| | - Frank C Ko
- Rush University Medical Center, Department of Internal Medicine, Division of Rheumatology, Chicago, IL USA
- Rush University Medical Center, Department of Anatomy & Cell Biology, Chicago, IL USA
| | - Anne-Marie Malfait
- Rush University Medical Center, Department of Internal Medicine, Division of Rheumatology, Chicago, IL USA
- Rush University Medical Center, Department of Anatomy & Cell Biology, Chicago, IL USA
| | - Rachel E Miller
- Rush University Medical Center, Department of Internal Medicine, Division of Rheumatology, Chicago, IL USA
- Rush University Medical Center, Department of Anatomy & Cell Biology, Chicago, IL USA
| |
Collapse
|
5
|
Feng SW, North TM, Wivell P, Pletcher A, Popratiloff A, Shibata M. Macrophages of multiple hematopoietic origins reside in the developing prostate. Development 2024; 151:dev203070. [PMID: 39082371 PMCID: PMC11385323 DOI: 10.1242/dev.203070] [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: 05/22/2024] [Accepted: 07/22/2024] [Indexed: 08/02/2024]
Abstract
Tissue-resident macrophages contribute to the organogenesis of many tissues. Growth of the prostate is regulated by androgens during puberty, yet androgens are considered immune suppressive. In this study, we characterized the localization, androgen receptor expression and hematopoietic origin of prostate macrophages, and transiently ablated macrophages during postnatal prostate organogenesis in the mouse. We show that myeloid cells were abundant in the prostate during puberty. However, nuclear androgen receptor expression was not detected in most macrophages. We found Cx3cr1, a marker for macrophages, monocytes and dendritic cells, expressed in interstitial macrophages surrounding the prostate and associated with nerve fibers. Furthermore, we provide evidence for the co-existence of embryonic origin, self-renewing, tissue-resident macrophages and recruited macrophages of bone-marrow monocyte origin in the prostate during puberty. Our findings suggest that prostate macrophages promote neural patterning and may shed further light on our understanding of the role of the innate immune system in prostate pathology in response to inflammation and in cancer.
Collapse
Affiliation(s)
- Sally W Feng
- Department of Anatomy and Cell Biology, The George Washington University School of Medicine and Health Sciences, Washington, DC 20052, USA
- The George Washington University Cancer Center, The George Washington University School of Medicine and Health Sciences, Washington, DC 20052, USA
| | - Tanya M North
- Department of Anatomy and Cell Biology, The George Washington University School of Medicine and Health Sciences, Washington, DC 20052, USA
- The George Washington University Cancer Center, The George Washington University School of Medicine and Health Sciences, Washington, DC 20052, USA
| | - Peri Wivell
- Department of Anatomy and Cell Biology, The George Washington University School of Medicine and Health Sciences, Washington, DC 20052, USA
- The George Washington University Cancer Center, The George Washington University School of Medicine and Health Sciences, Washington, DC 20052, USA
| | - Andrew Pletcher
- Department of Anatomy and Cell Biology, The George Washington University School of Medicine and Health Sciences, Washington, DC 20052, USA
- The George Washington University Cancer Center, The George Washington University School of Medicine and Health Sciences, Washington, DC 20052, USA
| | - Anastas Popratiloff
- Department of Anatomy and Cell Biology, The George Washington University School of Medicine and Health Sciences, Washington, DC 20052, USA
- GW Nanofabrication and Imaging Center, The George Washington University, Washington, DC 20052, USA
| | - Maho Shibata
- Department of Anatomy and Cell Biology, The George Washington University School of Medicine and Health Sciences, Washington, DC 20052, USA
- The George Washington University Cancer Center, The George Washington University School of Medicine and Health Sciences, Washington, DC 20052, USA
| |
Collapse
|
6
|
Ismahil MA, Zhou G, Gao M, Bansal SS, Patel B, Limdi N, Xie M, Antipenko S, Rokosh G, Hamid T, Prabhu SD. Splenic CD169 + Tim4 + Marginal Metallophilic Macrophages Are Essential for Wound Healing After Myocardial Infarction. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.09.24311769. [PMID: 39211861 PMCID: PMC11361232 DOI: 10.1101/2024.08.09.24311769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Fidelity of wound healing after myocardial infarction (MI) is an important determinant of subsequent adverse cardiac remodeling and failure. Macrophages derived from infiltrating Ly6C hi blood monocytes are a key component of this healing response; however, the importance of other macrophage populations is unclear. Here, using a variety of in vivo murine models and orthogonal approaches, including surgical myocardial infarction, splenectomy, parabiosis, cell adoptive transfer, lineage tracing and cell tracking, RNA sequencing, and functional characterization, we establish in mice an essential role for splenic CD169 + Tim4 + marginal metallophilic macrophages (MMMs) in post-MI wound healing. Splenic CD169 + Tim4 + MMMs circulate in blood as Ly6C low cells expressing macrophage markers and help populate CD169 + Tim4 + CCR2 - LYVE1 low macrophages in the naïve heart. After acute MI, splenic MMMs augment phagocytosis, CCR3 and CCR4 expression, and robustly mobilize to the heart, resulting in marked expansion of cardiac CD169 + Tim4 + LyVE1 low macrophages with an immunomodulatory and pro-resolving gene signature. These macrophages are obligatory for apoptotic neutrophil clearance, suppression of inflammation, and induction of a reparative macrophage phenotype in the infarcted heart. Splenic MMMs are both necessary and sufficient for post-MI wound healing, and limit late pathological remodeling. Liver X receptor-α agonist-induced expansion of the splenic marginal zone and MMMs during acute MI alleviates inflammation and improves short- and long-term cardiac remodeling. Finally, humans with acute ST-elevation MI also exhibit expansion of circulating CD169 + Tim4 + macrophages. We conclude that splenic CD169 + Tim4 + MMMs are required for pro-resolving and reparative responses after MI and can be manipulated for therapeutic benefit to limit long-term heart failure. CLINICAL PERSPECTIVE What is new?: We establish for the first time that metallophilic marginal macrophages (MMMs) from the spleen, expressing the markers CD169 and Tim4, circulate in blood and traffic to the heart to help maintain the CD169 + Tim4 + CCR2 - LYVE1 low macrophage population in the heart. After acute myocardial infarction, splenic MMMs augment cardiac trafficking in response to chemotactic signals, resulting in expansion of CD169 + Tim4 + macrophages in the heart that play an essential role in post-MI efferocytosis, wound healing and repair while limiting longer term adverse cardiac remodeling. Analogous to mice, humans also exhibit circulating CD169 + Tim4 + macrophages in the blood that expand after acute ST segment elevation MI. What are the clinical implications?: This study highlights the importance of the cardiosplenic axis in acute MI, and the splenic marginal zone, in determining the course and outcome of post-MI LV remodeling.Pharmacological expansion of splenic marginal zone macrophages alleviated post-MI adverse LV remodeling and inflammation, suggesting that splenic modulation is a potential translational therapeutic approach for limiting post-MI inflammation and improving heart repair.
Collapse
|
7
|
Liu Y, Liu X, Dorizas CA, Hao Z, Lee RK. Macrophages Modulate Optic Nerve Crush Injury Scar Formation and Retinal Ganglion Cell Function. Invest Ophthalmol Vis Sci 2024; 65:22. [PMID: 39140963 PMCID: PMC11328886 DOI: 10.1167/iovs.65.10.22] [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] [Indexed: 08/15/2024] Open
Abstract
Purpose Optic nerve (ON) injuries can result in vision loss via structural damage and cellular injury responses. Understanding the immune response, particularly the role of macrophages, in the cellular response to ON injury is crucial for developing therapeutic approaches which affect ON injury repair. The present study investigates the role of macrophages in ON injury response, fibrotic scar formation, and retinal ganglion cell (RGC) function. Methods The study utilizes macrophage Fas-induced apoptosis (MaFIA) mice to selectively deplete hematogenous macrophages and explores the impact macrophages have on ON injury responses. Histological and immunofluorescence analyses were used to evaluate macrophage expression levels and fibrotic scar formation. Pattern electroretinogram (PERG) recordings were used to assess RGC function as result of ON injury. Results Successful macrophage depletion was induced in MaFIA mice, which led to reduced fibrotic scar formation in the ON post-injury. Despite an increase in activated macrophages in the retina, RGC function was preserved, as demonstrated by normal PERG waveforms for up to 2 months post-injury. The study suggests a neuroprotective role for macrophage depletion in ON damage repair and highlights the complex immune response to ON injury. Conclusions To our knowledge, this study is the first to use MaFIA mice to demonstrate that targeted depletion of hematogenous macrophages leads to a significant reduction in scar size and the preservation of RGC functionality after ON injury. These findings highlight the key role of hematogenous macrophages in the response to ON injury and opens new avenues for therapeutic interventions in ON injuries. Future research should focus on investigating the distinct roles of macrophage subtypes in ON injury and potential macrophage-associated molecular targets to improve ON regeneration and repair.
Collapse
Affiliation(s)
- Yuan Liu
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, United States
| | - Xiangxiang Liu
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, United States
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Christopher A Dorizas
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, United States
| | - Zixuan Hao
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, United States
| | - Richard K Lee
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, United States
| |
Collapse
|
8
|
Chivers SB, Andrade MA, Hammack RJ, Shannonhouse J, Gomez R, Zhang Y, Nguyen B, Shah P, Kim YS, Toney GM, Jeske NA. Peripheral macrophages contribute to nociceptor priming in mice with chronic intermittent hypoxia. Sci Signal 2024; 17:eadn8936. [PMID: 39078919 PMCID: PMC11412124 DOI: 10.1126/scisignal.adn8936] [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: 01/05/2024] [Accepted: 06/13/2024] [Indexed: 09/21/2024]
Abstract
Obstructive sleep apnea (OSA) is a prevalent sleep disorder that is associated with increased incidence of chronic musculoskeletal pain. We investigated the mechanism of this association in a mouse model of chronic intermittent hypoxia (CIH) that mimics the repetitive hypoxemias of OSA. After 14 days of CIH, both male and female mice exhibited behaviors indicative of persistent pain, with biochemical markers in the spinal cord dorsal horn and sensory neurons of the dorsal root ganglia consistent with hyperalgesic priming. CIH, but not sleep fragmentation alone, induced an increase in macrophage recruitment to peripheral sensory tissues (sciatic nerve and dorsal root ganglia), an increase in inflammatory cytokines in the circulation, and nociceptor sensitization. Peripheral macrophage ablation blocked CIH-induced hyperalgesic priming. The findings suggest that correcting the hypoxia or targeting macrophage signaling might suppress persistent pain in patients with OSA.
Collapse
Affiliation(s)
- Samuel B. Chivers
- Department of Oral and Maxillofacial Surgery, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Mary Ann Andrade
- Department of Physiology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Robert J. Hammack
- Department of Physiology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - John Shannonhouse
- Department of Oral and Maxillofacial Surgery, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Ruben Gomez
- Department of Oral and Maxillofacial Surgery, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Yan Zhang
- Department of Oral and Maxillofacial Surgery, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Brian Nguyen
- Department of Oral and Maxillofacial Surgery, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Pankil Shah
- Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Yu Shin Kim
- Department of Oral and Maxillofacial Surgery, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Glenn M. Toney
- Department of Physiology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Nathaniel A. Jeske
- Department of Oral and Maxillofacial Surgery, University of Texas Health San Antonio, San Antonio, TX, USA
- Department of Physiology, University of Texas Health San Antonio, San Antonio, TX, USA
- Department of Pharmacology, University of Texas Health San Antonio, San Antonio, TX, USA
| |
Collapse
|
9
|
Cortez I, Gaffney CM, Vichare R, Crelli CV, Liu L, Lee E, Edralin J, Nichols JM, Pham HV, Mehdi S, Janjic JM, Shepherd AJ. Neuromuscular Polytrauma Pain is Resolved by Macrophage COX-2 Nanoimmunomodulation. Int J Nanomedicine 2024; 19:7253-7271. [PMID: 39050880 PMCID: PMC11268785 DOI: 10.2147/ijn.s460418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 07/10/2024] [Indexed: 07/27/2024] Open
Abstract
Soft tissue injuries often involve muscle and peripheral nerves and are qualitatively distinct from single-tissue injuries. Prior research suggests that damaged innervation compromises wound healing. To test this in a traumatic injury context, we developed a novel mouse model of nerve and lower limb polytrauma, which features greater pain hypersensitivity and more sustained macrophage infiltration than either injury in isolation. We also show that macrophages are crucial mediators of pain hypersensitivity in this model by delivering macrophage-targeted nanoemulsions laden with the cyclooxygenase-2 (COX-2) inhibitor celecoxib. This treatment was more effective in males than females, and more effective when delivered 3 days post-injury than 7 days post-injury. The COX-2 inhibiting nanoemulsion drove widespread anti-inflammatory changes in cytokine expression in polytrauma-affected peripheral nerves. Our data shed new light on the modulation of inflammation by injured nerve input and demonstrate macrophage-targeted nanoimmunomodulation can produce rapid and sustained pain relief following complex injuries.
Collapse
Affiliation(s)
- Ibdanelo Cortez
- Laboratories of Neuroimmunology, Department of Symptom Research, the University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Caitlyn M Gaffney
- Laboratories of Neuroimmunology, Department of Symptom Research, the University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Riddhi Vichare
- School of Pharmacy, Duquesne University, Pittsburgh, PA, 15282, USA
| | - Caitlin V Crelli
- School of Pharmacy, Duquesne University, Pittsburgh, PA, 15282, USA
| | - Lu Liu
- School of Pharmacy, Duquesne University, Pittsburgh, PA, 15282, USA
| | - Eric Lee
- Laboratories of Neuroimmunology, Department of Symptom Research, the University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jules Edralin
- Laboratories of Neuroimmunology, Department of Symptom Research, the University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - James M Nichols
- Laboratories of Neuroimmunology, Department of Symptom Research, the University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Hoang Vu Pham
- Laboratories of Neuroimmunology, Department of Symptom Research, the University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Syed Mehdi
- Laboratories of Neuroimmunology, Department of Symptom Research, the University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jelena M Janjic
- School of Pharmacy, Duquesne University, Pittsburgh, PA, 15282, USA
| | - Andrew J Shepherd
- Laboratories of Neuroimmunology, Department of Symptom Research, the University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| |
Collapse
|
10
|
Iglesias-Velazquez O, Gf Tresguerres F, F Tresguerres I, Leco-Berrocal I, Lopez-Pintor R, Baca L, Torres J. OsteoMac: A new player on the bone biology scene. Ann Anat 2024; 254:152244. [PMID: 38492654 DOI: 10.1016/j.aanat.2024.152244] [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: 12/05/2023] [Revised: 02/21/2024] [Accepted: 03/05/2024] [Indexed: 03/18/2024]
Abstract
The knowledge of bone biology has undergone major advances in recent decades. In bone, resorbing osteoclasts have classically been described as tissue-resident macrophages, however, it is currently known that a new subtype of macrophages, called OsteoMacs, are specialised bone-resident macrophages, which, depending on certain conditions, may play an important role not only in bone homeostasis, but also in promoting pro-anabolic functions or in creating an inflammatory environment. There is growing evidence that these osteal macrophages may influence the development of bone-loss diseases. It is essential to understand the biological bases underlying bone physiological processes to search for new therapeutic targets for bone-loss diseases, such as osteoporosis, rheumatoid arthritis, or even periodontal disease. This narrative review provides an update on the origin, characterisation, and possible roles of osteoMacs in bone biology. Finally, the potential clinical applications of this new cell in bone-loss disorders are discussed.
Collapse
Affiliation(s)
- Oscar Iglesias-Velazquez
- Department of Dental Clinical Specialties, Faculty of Dentistry, Complutense University of Madrid, Spain
| | - Francisco Gf Tresguerres
- Department of Dental Clinical Specialties, Faculty of Dentistry, Complutense University of Madrid, Spain
| | - Isabel F Tresguerres
- Department of Dental Clinical Specialties, Faculty of Dentistry, Complutense University of Madrid, Spain.
| | - Isabel Leco-Berrocal
- Department of Dental Clinical Specialties, Faculty of Dentistry, Complutense University of Madrid, Spain
| | - Rosa Lopez-Pintor
- Department of Dental Clinical Specialties, Faculty of Dentistry, Complutense University of Madrid, Spain
| | - Laura Baca
- Department of Dental Clinical Specialties, Faculty of Dentistry, Complutense University of Madrid, Spain
| | - Jesus Torres
- Department of Dental Clinical Specialties, Faculty of Dentistry, Complutense University of Madrid, Spain
| |
Collapse
|
11
|
Molitoris KH, Huang M, Baht GS. Osteoimmunology of Fracture Healing. Curr Osteoporos Rep 2024; 22:330-339. [PMID: 38616228 PMCID: PMC11186872 DOI: 10.1007/s11914-024-00869-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/26/2024] [Indexed: 04/16/2024]
Abstract
PURPOSE OF REVIEW The purpose of this review is to summarize what is known in the literature about the role inflammation plays during bone fracture healing. Bone fracture healing progresses through four distinct yet overlapping phases: formation of the hematoma, development of the cartilaginous callus, development of the bony callus, and finally remodeling of the fracture callus. Throughout this process, inflammation plays a critical role in robust bone fracture healing. RECENT FINDINGS At the onset of injury, vessel and matrix disruption lead to the generation of an inflammatory response: inflammatory cells are recruited to the injury site where they differentiate, activate, and/or polarize to secrete cytokines for the purposes of cell signaling and cell recruitment. This process is altered by age and by sex. Bone fracture healing is heavily influenced by the presence of inflammatory cells and cytokines within the healing tissue.
Collapse
Affiliation(s)
- Kristin Happ Molitoris
- Department of Orthopaedic Surgery, Duke Molecular Physiology Institute, Duke University, 300 North Duke Street, Durham, NC, 27701, USA
| | - Mingjian Huang
- Department of Orthopaedic Surgery, Duke Molecular Physiology Institute, Duke University, 300 North Duke Street, Durham, NC, 27701, USA
| | - Gurpreet Singh Baht
- Department of Orthopaedic Surgery, Duke Molecular Physiology Institute, Duke University, 300 North Duke Street, Durham, NC, 27701, USA.
| |
Collapse
|
12
|
Perzolli A, Koedijk JB, Zwaan CM, Heidenreich O. Targeting the innate immune system in pediatric and adult AML. Leukemia 2024; 38:1191-1201. [PMID: 38459166 PMCID: PMC11147779 DOI: 10.1038/s41375-024-02217-7] [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: 01/29/2024] [Revised: 02/27/2024] [Accepted: 02/29/2024] [Indexed: 03/10/2024]
Abstract
While the introduction of T cell-based immunotherapies has improved outcomes in many cancer types, the development of immunotherapies for both adult and pediatric AML has been relatively slow and limited. In addition to the need to identify suitable target antigens, a better understanding of the immunosuppressive tumor microenvironment is necessary for the design of novel immunotherapy approaches. To date, most immune characterization studies in AML have focused on T cells, while innate immune lineages such as monocytes, granulocytes and natural killer (NK) cells, received less attention. In solid cancers, studies have shown that innate immune cells, such as macrophages, myeloid-derived suppressor cells and neutrophils are highly plastic and may differentiate into immunosuppressive cells depending on signals received in their microenvironment, while NK cells appear to be functionally impaired. Hence, an in-depth characterization of the innate immune compartment in the TME is urgently needed to guide the development of immunotherapeutic interventions for AML. In this review, we summarize the current knowledge on the innate immune compartment in AML, and we discuss how targeting its components may enhance T cell-based- and other immunotherapeutic approaches.
Collapse
Affiliation(s)
- Alicia Perzolli
- Princess Máxima Center for Pediatric Oncology, 3584 CS, Utrecht, The Netherlands
- Department of Pediatric Oncology, Erasmus MC/Sophia Children's Hospital, 3015 GD, Rotterdam, The Netherlands
| | - Joost B Koedijk
- Princess Máxima Center for Pediatric Oncology, 3584 CS, Utrecht, The Netherlands
- Department of Pediatric Oncology, Erasmus MC/Sophia Children's Hospital, 3015 GD, Rotterdam, The Netherlands
| | - C Michel Zwaan
- Princess Máxima Center for Pediatric Oncology, 3584 CS, Utrecht, The Netherlands
- Department of Pediatric Oncology, Erasmus MC/Sophia Children's Hospital, 3015 GD, Rotterdam, The Netherlands
| | - Olaf Heidenreich
- Princess Máxima Center for Pediatric Oncology, 3584 CS, Utrecht, The Netherlands.
- Wolfson Childhood Cancer Research Centre, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK.
| |
Collapse
|
13
|
Dourson AJ, Fadaka AO, Warshak AM, Paranjpe A, Weinhaus B, Queme LF, Hofmann MC, Evans HM, Donmez OA, Forney C, Weirauch MT, Kottyan LC, Lucas D, Deepe GS, Jankowski MP. Macrophage memories of early-life injury drive neonatal nociceptive priming. Cell Rep 2024; 43:114129. [PMID: 38640063 PMCID: PMC11197107 DOI: 10.1016/j.celrep.2024.114129] [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: 02/14/2023] [Revised: 01/05/2024] [Accepted: 04/04/2024] [Indexed: 04/21/2024] Open
Abstract
The developing peripheral nervous and immune systems are functionally distinct from those of adults. These systems are vulnerable to early-life injury, which influences outcomes related to nociception following subsequent injury later in life (i.e., "neonatal nociceptive priming"). The underpinnings of this phenomenon are unclear, although previous work indicates that macrophages are trained by inflammation and injury. Our findings show that macrophages are both necessary and partially sufficient to drive neonatal nociceptive priming, possibly due to a long-lasting remodeling in chromatin structure. The p75 neurotrophic factor receptor is an important effector in regulating neonatal nociceptive priming through modulation of the inflammatory profile of rodent and human macrophages. This "pain memory" is long lasting in females and can be transferred to a naive host to alter sex-specific pain-related behaviors. This study reveals a mechanism by which acute, neonatal post-surgical pain drives a peripheral immune-related predisposition to persistent pain following a subsequent injury.
Collapse
Affiliation(s)
- Adam J Dourson
- Department of Anesthesia, Division of Pain Management, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Adewale O Fadaka
- Department of Anesthesia, Division of Pain Management, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Anna M Warshak
- Department of Anesthesia, Division of Pain Management, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Aditi Paranjpe
- Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Benjamin Weinhaus
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Medical Center, Cincinnati, OH, USA
| | - Luis F Queme
- Department of Anesthesia, Division of Pain Management, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Megan C Hofmann
- Department of Anesthesia, Division of Pain Management, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Heather M Evans
- Division of Infectious Diseases, University of Cincinnati, Cincinnati, OH, USA
| | - Omer A Donmez
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Carmy Forney
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Matthew T Weirauch
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Divisions of Biomedical Informatics and Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Leah C Kottyan
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Daniel Lucas
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Medical Center, Cincinnati, OH, USA; Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH 45229, USA
| | - George S Deepe
- Division of Infectious Diseases, Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Michael P Jankowski
- Department of Anesthesia, Division of Pain Management, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH 45229, USA; Pediatric Pain Research Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| |
Collapse
|
14
|
Billman ZP, Kovacs SB, Wei B, Kang K, Cissé OH, Miao EA. Caspase-1 activates gasdermin A in non-mammals. eLife 2024; 12:RP92362. [PMID: 38497531 PMCID: PMC10948149 DOI: 10.7554/elife.92362] [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: 03/19/2024] Open
Abstract
Gasdermins oligomerize to form pores in the cell membrane, causing regulated lytic cell death called pyroptosis. Mammals encode five gasdermins that can trigger pyroptosis: GSDMA, B, C, D, and E. Caspase and granzyme proteases cleave the linker regions of and activate GSDMB, C, D, and E, but no endogenous activation pathways are yet known for GSDMA. Here, we perform a comprehensive evolutionary analysis of the gasdermin family. A gene duplication of GSDMA in the common ancestor of caecilian amphibians, reptiles, and birds gave rise to GSDMA-D in mammals. Uniquely in our tree, amphibian, reptile, and bird GSDMA group in a separate clade than mammal GSDMA. Remarkably, GSDMA in numerous bird species contain caspase-1 cleavage sites like YVAD or FASD in the linker. We show that GSDMA from birds, amphibians, and reptiles are all cleaved by caspase-1. Thus, GSDMA was originally cleaved by the host-encoded protease caspase-1. In mammals the caspase-1 cleavage site in GSDMA is disrupted; instead, a new protein, GSDMD, is the target of caspase-1. Mammal caspase-1 uses exosite interactions with the GSDMD C-terminal domain to confer the specificity of this interaction, whereas we show that bird caspase-1 uses a stereotypical tetrapeptide sequence to confer specificity for bird GSDMA. Our results reveal an evolutionarily stable association between caspase-1 and the gasdermin family, albeit a shifting one. Caspase-1 repeatedly changes its target gasdermin over evolutionary time at speciation junctures, initially cleaving GSDME in fish, then GSDMA in amphibians/reptiles/birds, and finally GSDMD in mammals.
Collapse
Affiliation(s)
- Zachary Paul Billman
- Department of Integrative Immunobiology; Molecular Genetics and Microbiology; Pathology; and Cell Biology, Duke University School of MedicineDurhamUnited States
- Department of Microbiology and Immunology, University of North Carolina at Chapel HillChapel HillUnited States
| | - Stephen Bela Kovacs
- Department of Integrative Immunobiology; Molecular Genetics and Microbiology; Pathology; and Cell Biology, Duke University School of MedicineDurhamUnited States
- Department of Microbiology and Immunology, University of North Carolina at Chapel HillChapel HillUnited States
| | - Bo Wei
- Department of Integrative Immunobiology; Molecular Genetics and Microbiology; Pathology; and Cell Biology, Duke University School of MedicineDurhamUnited States
| | - Kidong Kang
- Department of Integrative Immunobiology; Molecular Genetics and Microbiology; Pathology; and Cell Biology, Duke University School of MedicineDurhamUnited States
| | - Ousmane H Cissé
- Critical Care Medicine Department, National Institutes of Health Clinical CenterBethesdaUnited States
| | - Edward A Miao
- Department of Integrative Immunobiology; Molecular Genetics and Microbiology; Pathology; and Cell Biology, Duke University School of MedicineDurhamUnited States
| |
Collapse
|
15
|
Eugenín J, Beltrán-Castillo S, Irribarra E, Pulgar-Sepúlveda R, Abarca N, von Bernhardi R. Microglial reactivity in brainstem chemosensory nuclei in response to hypercapnia. Front Physiol 2024; 15:1332355. [PMID: 38476146 PMCID: PMC10927973 DOI: 10.3389/fphys.2024.1332355] [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: 11/02/2023] [Accepted: 02/08/2024] [Indexed: 03/14/2024] Open
Abstract
Microglia, the resident immune cells of the CNS, surveil, detect, and respond to various extracellular signals. Depending on the nature of these signals, an integrative microglial response can be triggered, resulting in a phenotypic transformation. Here, we evaluate whether hypercapnia modifies microglia phenotype in brainstem respiratory-related nuclei. Adult C57BL/6 inbred mice were exposed to 10% CO2 enriched air (hypercapnia), or pure air (control), for 10 or 30 min and immediately processed for immunohistochemistry to detect the ubiquitous microglia marker, ionized calcium binding adaptor molecule 1 (Iba1). Hypercapnia for thirty, but not 10 min reduced the Iba1 labeling percent coverage in the ventral respiratory column (VRC), raphe nucleus (RN), and nucleus tractus solitarius (NTS) and the number of primary branches in VRC. The morphological changes persisted, at least, for 60 min breathing air after the hypercapnic challenge. No significant changes were observed in Iba1+ cells in the spinal trigeminal nucleus (Sp5) and the hippocampus. In CF-1 outbred mice, 10% CO2 followed by 60 min of breathing air, resulted in the reduction of Iba1 labeling percent coverage and the number and length of primary branches in VRC, RN, and NTS. No morphological change was observed in Iba1+ cells in Sp5 and hippocampus. Double immunofluorescence revealed that prolonged hypercapnia increased the expression of CD86, an inflammatory marker for reactive state microglia, in Iba1+ cells in VRC, RN, and NTS, but not in Sp5 and hippocampus in CF-1 mice. By contrast, the expression of CD206, a marker of regulatory state microglia, persisted unmodified. In brainstem, but not in hippocampal microglia cultures, hypercapnia increased the level of IL1β, but not that of TGFβ measured by ELISA. Our results show that microglia from respiratory-related chemosensory nuclei, are reactive to prolonged hypercapnia acquiring an inflammatory-like phenotype.
Collapse
Affiliation(s)
- Jaime Eugenín
- Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Sebastián Beltrán-Castillo
- Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
- Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O’Higgins, Santiago, Chile
| | - Estefanía Irribarra
- Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | | | - Nicolás Abarca
- Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Rommy von Bernhardi
- Facultad de Odontología y Ciencias de la Rehabilitación, Universidad San Sebastián, Santiago, Chile
| |
Collapse
|
16
|
Capobianco CA, Hankenson KD, Knights AJ. Temporal dynamics of immune-stromal cell interactions in fracture healing. Front Immunol 2024; 15:1352819. [PMID: 38455063 PMCID: PMC10917940 DOI: 10.3389/fimmu.2024.1352819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 02/06/2024] [Indexed: 03/09/2024] Open
Abstract
Bone fracture repair is a complex, multi-step process that involves communication between immune and stromal cells to coordinate the repair and regeneration of damaged tissue. In the US, 10% of all bone fractures do not heal properly without intervention, resulting in non-union. Complications from non-union fractures are physically and financially debilitating. We now appreciate the important role that immune cells play in tissue repair, and the necessity of the inflammatory response in initiating healing after skeletal trauma. The temporal dynamics of immune and stromal cell populations have been well characterized across the stages of fracture healing. Recent studies have begun to untangle the intricate mechanisms driving the immune response during normal or atypical, delayed healing. Various in vivo models of fracture healing, including genetic knockouts, as well as in vitro models of the fracture callus, have been implemented to enable experimental manipulation of the heterogeneous cellular environment. The goals of this review are to (1): summarize our current understanding of immune cell involvement in fracture healing (2); describe state-of-the art approaches to study inflammatory cells in fracture healing, including computational and in vitro models; and (3) identify gaps in our knowledge concerning immune-stromal crosstalk during bone healing.
Collapse
Affiliation(s)
- Christina A. Capobianco
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, United States
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Kurt D. Hankenson
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, United States
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Alexander J. Knights
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
17
|
Miron RJ, Bohner M, Zhang Y, Bosshardt DD. Osteoinduction and osteoimmunology: Emerging concepts. Periodontol 2000 2024; 94:9-26. [PMID: 37658591 DOI: 10.1111/prd.12519] [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: 05/14/2023] [Revised: 06/23/2023] [Accepted: 07/20/2023] [Indexed: 09/03/2023]
Abstract
The recognition and importance of immune cells during bone regeneration, including around bone biomaterials, has led to the development of an entire field termed "osteoimmunology," which focuses on the connection and interplay between the skeletal system and immune cells. Most studies have focused on the "osteogenic" capacity of various types of bone biomaterials, and much less focus has been placed on immune cells despite being the first cell type in contact with implantable devices. Thus, the amount of literature generated to date on this topic makes it challenging to extract needed information. This review article serves as a guide highlighting advancements made in the field of osteoimmunology emphasizing the role of the osteoimmunomodulatory properties of biomaterials and their impact on osteoinduction. First, the various immune cell types involved in bone biomaterial integration are discussed, including the prominent role of osteal macrophages (OsteoMacs) during bone regeneration. Thereafter, key biomaterial properties, including topography, wettability, surface charge, and adsorption of cytokines, growth factors, ions, and other bioactive molecules, are discussed in terms of their impact on immune responses. These findings highlight and recognize the importance of the immune system and osteoimmunology, leading to a shift in the traditional models used to understand and evaluate biomaterials for bone regeneration.
Collapse
Affiliation(s)
- Richard J Miron
- Department of Periodontology, University of Bern, Bern, Switzerland
| | | | - Yufeng Zhang
- Department of Oral Implantology, University of Wuhan, Wuhan, China
| | | |
Collapse
|
18
|
Billman ZP, Kovacs SB, Wei B, Kang K, Cissé OH, Miao EA. Caspase-1 activates gasdermin A in non-mammals. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.28.559989. [PMID: 37987010 PMCID: PMC10659411 DOI: 10.1101/2023.09.28.559989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Gasdermins oligomerize to form pores in the cell membrane, causing regulated lytic cell death called pyroptosis. Mammals encode five gasdermins that can trigger pyroptosis: GSDMA, B, C, D, and E. Caspase and granzyme proteases cleave the linker regions of and activate GSDMB, C, D, and E, but no endogenous activation pathways are yet known for GSDMA. Here, we perform a comprehensive evolutionary analysis of the gasdermin family. A gene duplication of GSDMA in the common ancestor of caecilian amphibians, reptiles and birds gave rise to GSDMA-D in mammals. Uniquely in our tree, amphibian, reptile and bird GSDMA group in a separate clade than mammal GSDMA. Remarkably, GSDMA in numerous bird species contain caspase-1 cleavage sites like YVAD or FASD in the linker. We show that GSDMA from birds, amphibians, and reptiles are all cleaved by caspase-1. Thus, GSDMA was originally cleaved by the host-encoded protease caspase-1. In mammals the caspase-1 cleavage site in GSDMA is disrupted; instead, a new protein, GSDMD, is the target of caspase-1. Mammal caspase-1 uses exosite interactions with the GSDMD C-terminal domain to confer the specificity of this interaction, whereas we show that bird caspase-1 uses a stereotypical tetrapeptide sequence to confer specificity for bird GSDMA. Our results reveal an evolutionarily stable association between caspase-1 and the gasdermin family, albeit a shifting one. Caspase-1 repeatedly changes its target gasdermin over evolutionary time at speciation junctures, initially cleaving GSDME in fish, then GSDMA in amphibians/reptiles/birds, and finally GSDMD in mammals.
Collapse
Affiliation(s)
- Zachary P Billman
- Duke University School of Medicine
- National Institutes of Health University of North Carolina at Chapel Hill
- Departments of: Integrative Immunobiology; Molecular Genetics and Microbiology; Cell Biology; Pathology; Durham, NC, USA
- Department of Microbiology and Immunology; Chapel Hill, NC, USA
| | - Stephen B Kovacs
- Duke University School of Medicine
- National Institutes of Health University of North Carolina at Chapel Hill
- Departments of: Integrative Immunobiology; Molecular Genetics and Microbiology; Cell Biology; Pathology; Durham, NC, USA
- Department of Microbiology and Immunology; Chapel Hill, NC, USA
| | - Bo Wei
- Duke University School of Medicine
- Departments of: Integrative Immunobiology; Molecular Genetics and Microbiology; Cell Biology; Pathology; Durham, NC, USA
| | - Kidong Kang
- Duke University School of Medicine
- Departments of: Integrative Immunobiology; Molecular Genetics and Microbiology; Cell Biology; Pathology; Durham, NC, USA
| | - Ousmane H Cissé
- National Institutes of Health
- Critical Care Medicine Department; Bethesda, MD, USA
| | - Edward A Miao
- Duke University School of Medicine
- National Institutes of Health University of North Carolina at Chapel Hill
- Departments of: Integrative Immunobiology; Molecular Genetics and Microbiology; Cell Biology; Pathology; Durham, NC, USA
| |
Collapse
|
19
|
Dourson AJ, Fadaka AO, Warshak AM, Paranjpe A, Weinhaus B, Queme LF, Hofmann MC, Evans HM, Donmez OA, Forney C, Weirauch MT, Kottyan LT, Lucas D, Deepe GS, Jankowski MP. Macrophage epigenetic memories of early life injury drive neonatal nociceptive priming. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.13.528015. [PMID: 36824978 PMCID: PMC9948986 DOI: 10.1101/2023.02.13.528015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
The developing peripheral nervous and immune systems are functionally distinct from adults. These systems are vulnerable to early life injury, which influences outcomes related to nociception following subsequent injury later in life (neonatal nociceptive priming). The underpinnings of this phenomenon are largely unknown, although previous work indicates that macrophages are epigenetically trained by inflammation and injury. We found that macrophages are both necessary and partially sufficient to drive neonatal nociceptive priming possibly due to a long-lasting epigenetic remodeling. The p75 neurotrophic factor receptor (NTR) was an important effector in regulating neonatal nociceptive priming through modulation of the inflammatory profile of rodent and human macrophages. This pain memory was long lasting in females and could be transferred to a naive host to alter sex-specific pain-related behaviors. This study reveals a novel mechanism by which acute, neonatal post-surgical pain drives a peripheral immune-related predisposition to persistent pain following a subsequent injury.
Collapse
|
20
|
Bhattacharyya ND, Kyaw W, McDonald MM, Dhenni R, Grootveld AK, Xiao Y, Chai R, Khoo WH, Danserau LC, Sergio CM, Timpson P, Lee WM, Croucher PI, Phan TG. Minimally invasive longitudinal intravital imaging of cellular dynamics in intact long bone. Nat Protoc 2023; 18:3856-3880. [PMID: 37857852 DOI: 10.1038/s41596-023-00894-9] [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: 01/16/2023] [Accepted: 07/28/2023] [Indexed: 10/21/2023]
Abstract
Intravital two-photon microscopy enables deep-tissue imaging at high temporospatial resolution in live animals. However, the endosteal bone compartment and underlying bone marrow pose unique challenges to optical imaging as light is absorbed, scattered and dispersed by thick mineralized bone matrix and the adipose-rich bone marrow. Early bone intravital imaging methods exploited gaps in the cranial sutures to bypass the need to penetrate through cortical bone. More recently, investigators have developed invasive methods to thin the cortical bone or implant imaging windows to image cellular dynamics in weight-bearing long bones. Here, we provide a step-by-step procedure for the preparation of animals for minimally invasive, nondestructive, longitudinal intravital imaging of the murine tibia. This method involves the use of mixed bone marrow radiation chimeras to unambiguously double-label osteoclasts and osteomorphs. The tibia is exposed by a simple skin incision and an imaging chamber constructed using thermoconductive T-putty. Imaging sessions up to 12 h long can be repeated over multiple timepoints to provide a longitudinal time window into the endosteal and marrow niches. The approach can be used to investigate cellular dynamics in bone remodeling, cancer cell life cycle and hematopoiesis, as well as long-lived humoral and cellular immunity. The procedure requires an hour to complete and is suitable for users with minimal prior expertise in small animal surgery.
Collapse
Affiliation(s)
- Nayan Deger Bhattacharyya
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
| | - Wunna Kyaw
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
| | - Michelle M McDonald
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Rama Dhenni
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
| | - Abigail K Grootveld
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
| | - Ya Xiao
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
| | - Ryan Chai
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
| | - Weng Hua Khoo
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
| | - Linda C Danserau
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
- ACRF INCITe Centre for Intravital Imaging of Niches for Cancer Immune Therapy, Sydney, New South Wales, Australia
| | - C Marcelo Sergio
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Paul Timpson
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
- ACRF INCITe Centre for Intravital Imaging of Niches for Cancer Immune Therapy, Sydney, New South Wales, Australia
| | - Woei Ming Lee
- ACRF INCITe Centre for Intravital Imaging of Niches for Cancer Immune Therapy, Sydney, New South Wales, Australia
- John Curtin School of Medical Research, Australian National University, Canberra, New South Wales, Australia
| | - Peter I Croucher
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
- ACRF INCITe Centre for Intravital Imaging of Niches for Cancer Immune Therapy, Sydney, New South Wales, Australia
| | - Tri Giang Phan
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia.
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia.
- ACRF INCITe Centre for Intravital Imaging of Niches for Cancer Immune Therapy, Sydney, New South Wales, Australia.
| |
Collapse
|
21
|
Guo D, Jurek R, Beaumont KA, Sharp DS, Tan SY, Mariana A, Failes TW, Grootveld AK, Bhattacharyya ND, Phan TG, Arndt GM, Jain R, Weninger W, Tikoo S. Invasion-Block and S-MARVEL: A high-content screening and image analysis platform identifies ATM kinase as a modulator of melanoma invasion and metastasis. Proc Natl Acad Sci U S A 2023; 120:e2303978120. [PMID: 37963252 PMCID: PMC10666109 DOI: 10.1073/pnas.2303978120] [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: 03/09/2023] [Accepted: 08/13/2023] [Indexed: 11/16/2023] Open
Abstract
Robust high-throughput assays are crucial for the effective functioning of a drug discovery pipeline. Herein, we report the development of Invasion-Block, an automated high-content screening platform for measuring invadopodia-mediated matrix degradation as a readout for the invasive capacity of cancer cells. Combined with Smoothen-Mask and Reveal, a custom-designed, automated image analysis pipeline, this platform allowed us to evaluate melanoma cell invasion capacity posttreatment with two libraries of compounds comprising 3840 U.S. Food and Drug Administration (FDA)-approved drugs with well-characterized safety and bioavailability profiles in humans as well as a kinase inhibitor library comprising 210 biologically active compounds. We found that Abl/Src, PKC, PI3K, and Ataxia-telangiectasia mutated (ATM) kinase inhibitors significantly reduced melanoma cell invadopodia formation and cell invasion. Abrogation of ATM expression in melanoma cells via CRISPR-mediated gene knockout reduced 3D invasion in vitro as well as spontaneous lymph node metastasis in vivo. Together, this study established a rapid screening assay coupled with a customized image-analysis pipeline for the identification of antimetastatic drugs. Our study implicates that ATM may serve as a potent therapeutic target for the treatment of melanoma cell spread in patients.
Collapse
Affiliation(s)
- Dajiang Guo
- Immune Imaging Program, Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW2050, Australia
- Sydney Medical School, The University of Sydney, Camperdown, NSW2050, Australia
| | - Russell Jurek
- Australia Telescope National Facility, The Commonwealth Scientific and Industrial Research Organisation (CSIRO) Astronomy and Space Science, Australia Telescope National Facility, MarsfieldNSW2122, Australia
| | - Kimberley A. Beaumont
- Immune Imaging Program, Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW2050, Australia
- Sydney Medical School, The University of Sydney, Camperdown, NSW2050, Australia
| | - Danae S. Sharp
- Immune Imaging Program, Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW2050, Australia
| | - Sioh-Yang Tan
- Immune Imaging Program, Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW2050, Australia
| | - Anna Mariana
- The Australian Cancer Research Foundation (ACRF) Drug Discovery Centre for Childhood Cancer, Children’s Cancer Institute, Lowy Cancer Research Centre, University of New South Wales (UNSW) Sydney, Sydney, NSW2052, Australia
| | - Timothy W. Failes
- The Australian Cancer Research Foundation (ACRF) Drug Discovery Centre for Childhood Cancer, Children’s Cancer Institute, Lowy Cancer Research Centre, University of New South Wales (UNSW) Sydney, Sydney, NSW2052, Australia
- School of Clinical Medicine, UNSW Medicine and Health, University of New South Wales (UNSW) Sydney, Sydney, NSW2052, Australia
| | - Abigail K. Grootveld
- Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW2010, Australia
- St Vincent’s Healthcare Clinical Campus, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW2010, Australia
| | - Nayan D. Bhattacharyya
- Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW2010, Australia
- St Vincent’s Healthcare Clinical Campus, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW2010, Australia
| | - Tri Giang Phan
- Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW2010, Australia
- St Vincent’s Healthcare Clinical Campus, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW2010, Australia
| | - Greg M. Arndt
- The Australian Cancer Research Foundation (ACRF) Drug Discovery Centre for Childhood Cancer, Children’s Cancer Institute, Lowy Cancer Research Centre, University of New South Wales (UNSW) Sydney, Sydney, NSW2052, Australia
- School of Clinical Medicine, UNSW Medicine and Health, University of New South Wales (UNSW) Sydney, Sydney, NSW2052, Australia
| | - Rohit Jain
- Immune Imaging Program, Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW2050, Australia
- Sydney Medical School, The University of Sydney, Camperdown, NSW2050, Australia
- Department of Dermatology, Medical University of Vienna, Vienna1090, Austria
| | - Wolfgang Weninger
- Immune Imaging Program, Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW2050, Australia
- Sydney Medical School, The University of Sydney, Camperdown, NSW2050, Australia
- Department of Dermatology, Medical University of Vienna, Vienna1090, Austria
| | - Shweta Tikoo
- Immune Imaging Program, Centenary Institute, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW2050, Australia
- Sydney Medical School, The University of Sydney, Camperdown, NSW2050, Australia
- Department of Dermatology, Medical University of Vienna, Vienna1090, Austria
| |
Collapse
|
22
|
Bousso P, Grandjean CL. Immunomodulation under the lens of real-time in vivo imaging. Eur J Immunol 2023; 53:e2249921. [PMID: 37051691 DOI: 10.1002/eji.202249921] [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: 07/27/2022] [Revised: 11/16/2022] [Accepted: 04/11/2023] [Indexed: 04/14/2023]
Abstract
Modulation of cells and molecules of the immune system not only represents a major opportunity to treat a variety of diseases including infections, cancer, autoimmune, and inflammatory disorders but could also help understand the intricacies of immune responses. A detailed mechanistic understanding of how a specific immune intervention may provide clinical benefit is essential for the rational design of efficient immunomodulators. Visualizing the impact of immunomodulation in real-time and in vivo has emerged as an important approach to achieve this goal. In this review, we aim to illustrate how multiphoton intravital imaging has helped clarify the mode of action of immunomodulatory strategies such as antibodies or cell therapies. We also discuss how optogenetics combined with imaging will further help manipulate and precisely understand immunomodulatory pathways. Combined with other single-cell technologies, in vivo dynamic imaging has therefore a major potential for guiding preclinical development of immunomodulatory drugs.
Collapse
Affiliation(s)
- Philippe Bousso
- Dynamics of Immune Responses Unit, Institut Pasteur, INSERM U1223, Université de Paris Cité, Paris, France
| | - Capucine L Grandjean
- Dynamics of Immune Responses Unit, Institut Pasteur, INSERM U1223, Université de Paris Cité, Paris, France
| |
Collapse
|
23
|
Zhang Y, Ji Q. Macrophage polarization in osteoarthritis progression: a promising therapeutic target. Front Cell Dev Biol 2023; 11:1269724. [PMID: 37954210 PMCID: PMC10639142 DOI: 10.3389/fcell.2023.1269724] [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: 07/30/2023] [Accepted: 10/16/2023] [Indexed: 11/14/2023] Open
Abstract
Osteoarthritis (OA) is one of the leading causes of pain and disability in the elderly. Synovitis, cartilage destruction and osteophyte formation histologically manifest OA. Unfortunately, there is currently no effective therapy to delay its progression and the underlying mechanisms of OA require further exploration. Macrophage is a main cellular component of joint synovium. It is highly plastic and can be stimulated to polarize to different phenotypes, namely, the pro-inflammatory phenotype (M1) and the anti-inflammatory/tissue-repairing phenotype (M2). Ample evidence has demonstrated the vital roles of macrophages in the progression of OA. Imbalanced M1/M2 ratio is significantly related to OA severity indicating macrophage polarization might be a promising therapeutic target for OA. In this review, we summarized the involvements of polarized macrophages in synovitis, cartilage degradation, osteophyte formation and OA-related chronic pain. Promising therapies targeting macrophage polarization including the intra-articular cell/derivates-based therapy and the alternative non-invasive intervention such as photobiomodulation therapy were reviewed as well.
Collapse
Affiliation(s)
| | - Quanbo Ji
- Department of Orthopedics, The General Hospital of Chinese PLA, Beijing, China
| |
Collapse
|
24
|
Bahri M, Anstee JE, Opzoomer JW, Arnold JN. Perivascular tumor-associated macrophages and their role in cancer progression. Essays Biochem 2023; 67:919-928. [PMID: 37199172 PMCID: PMC10539944 DOI: 10.1042/ebc20220242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 05/19/2023]
Abstract
Perivascular (Pv) tumor-associated macrophages (TAMs) are a highly specialized stromal subset within the tumor microenvironment (TME) that are defined by their spatial proximity, within one cell thickness, to blood vasculature. PvTAMs have been demonstrated to support a variety of pro-tumoral functions including angiogenesis, metastasis, and modulating the immune and stromal landscape. Furthermore, PvTAMs can also limit the response of anti-cancer and anti-angiogenic therapies and support tumor recurrence post-treatment. However, their role may not exclusively be pro-tumoral as PvTAMs can also have immune-stimulatory capabilities. PvTAMs are derived from a monocyte progenitor that develop and localize to the Pv niche as part of a multistep process which relies on a series of signals from tumor, endothelial and Pv mesenchymal cell populations. These cellular communications and signals create a highly specialized TAM subset that can also form CCR5-dependent multicellular 'nest' structures in the Pv niche. This review considers our current understanding of the role of PvTAMs, their markers for identification, development, and function in cancer. The role of PvTAMs in supporting disease progression and modulating the outcome from anti-cancer therapies highlight these cells as a therapeutic target. However, their resistance to pan-TAM targeting therapies, such as those targeting the colony stimulating factor-1 (CSF1)-CSF1 receptor axis, prompts the need for more targeted therapeutic approaches to be considered for this subset. This review highlights potential therapeutic strategies to target and modulate PvTAM development and function in the TME.
Collapse
Affiliation(s)
- Meriem Bahri
- School of Cancer and Pharmaceutical Sciences, King's College London, Faculty of Life Sciences and Medicine, Guy's Hospital, London SE1 1UL, United Kingdom
| | - Joanne E Anstee
- School of Cancer and Pharmaceutical Sciences, King's College London, Faculty of Life Sciences and Medicine, Guy's Hospital, London SE1 1UL, United Kingdom
| | - James W Opzoomer
- School of Cancer and Pharmaceutical Sciences, King's College London, Faculty of Life Sciences and Medicine, Guy's Hospital, London SE1 1UL, United Kingdom
| | - James N Arnold
- School of Cancer and Pharmaceutical Sciences, King's College London, Faculty of Life Sciences and Medicine, Guy's Hospital, London SE1 1UL, United Kingdom
| |
Collapse
|
25
|
Maisuria R, Norton A, Shao C, Bradley EW, Mansky K. Conditional Loss of MEF2C Expression in Osteoclasts Leads to a Sex-Specific Osteopenic Phenotype. Int J Mol Sci 2023; 24:12686. [PMID: 37628864 PMCID: PMC10454686 DOI: 10.3390/ijms241612686] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 08/03/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Myocyte enhancement factor 2C (MEF2C) is a transcription factor studied in the development of skeletal and smooth muscles. Bone resorption studies have exhibited that the reduced expression of MEF2C contributes to osteopetrosis and the dysregulation of pathological bone remodeling. Our current study aims to determine how MEF2C contributes to osteoclast differentiation and to analyze the skeletal phenotype of Mef2c-cKO mice (Cfms-cre; Mef2cfl/fl). qRT-PCR and Western blot demonstrated that Mef2c expression is highest during the early days of osteoclast differentiation. Osteoclast genes, including c-Fos, c-Jun, Dc-stamp, Cathepsin K, and Nfatc1, had a significant reduction in expression, along with a reduction in osteoclast size. Despite reduced CTX activity, female Mef2c cKO mice were osteopenic, with decreased bone formation as determined via a P1NP ELISA, and a reduced number of osteoblasts. There was no difference between male WT and Mef2c-cKO mice. Our results suggest that Mef2c is critical for osteoclastogenesis, and that its dysregulation leads to a sex-specific osteopenic phenotype.
Collapse
Affiliation(s)
- Ravi Maisuria
- Department of Developmental and Surgical Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN 55455, USA; (R.M.); (A.N.)
| | - Andrew Norton
- Department of Developmental and Surgical Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN 55455, USA; (R.M.); (A.N.)
| | - Cynthia Shao
- College of Biological Sciences, University of Minnesota, Minneapolis, MN 55455, USA;
| | - Elizabeth W. Bradley
- Department of Orthopedics, School of Medicine and Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA;
| | - Kim Mansky
- Department of Developmental and Surgical Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN 55455, USA; (R.M.); (A.N.)
| |
Collapse
|
26
|
Xiao L, Matharoo J, Chi J, Ma J, Chen M, Manley B, Xu P, Shi W, Felder RA, Sung SSJ, Jin L, Li X. Transient depletion of macrophages alters local inflammatory response at the site of disc herniation in a transgenic mouse model. Osteoarthritis Cartilage 2023; 31:894-907. [PMID: 36754251 PMCID: PMC10272080 DOI: 10.1016/j.joca.2023.01.574] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 10/06/2022] [Accepted: 01/10/2023] [Indexed: 02/10/2023]
Abstract
OBJECTIVE Macrophages are abundantly detected at sites of disc herniation, however, their function in the disease progression is unclear. We aim to investigate the functions of macrophages in acute disc herniation using a macrophage Fas-induced apoptosis (MaFIA) transgenic mouse strain. METHOD To transiently deplete macrophages, a dimerizer, AP20187, or vehicle solution was administered via intraperitoneal injection to MaFIA mice immediately, day 1 and 2 after annular puncture induced disc herniation. Local infiltrated tissues at disc hernia and DRGs at corresponding levels were harvested to analyze immune cells and neuroinflammation on postoperative day (POD) 6 by flow cytometry and/or immunostaining. Mouse spines were harvested to analyze structures of degenerated discs and adjacent vertebrae and to assess osteoclast activity by histology and tartrate-resistant acid phosphatase (TRAP) staining on POD 6, 13, and 20, respectively. RESULTS On POD 6, abundant macrophages were confirmed at disc hernia sites. Compared to vehicle control, AP20187 significantly reduced GFP+ cells in blood, spleen, and local inflammatory tissue. At disc hernia sites, AP20187 markedly reduced macrophages (CD11b+, F4/80+, GFP+CD11b+, CD11b+F4/80+) while increasing neutrophils and B cells. Transient macrophage depletion decreased ectopic bone formation and osteoclast activity in herniated discs and adjacent cortical bones for up to 20 days post herniation. Disc herniation elevated expressions of TNF-α, IL-6, substance P, calcitonin gene-related peptide, accompanied by increasing GFP+, CD11b+ and F4/80+ macrophages. Macrophage depletion did not attenuate these markers of neuroinflammation. CONCLUSIONS Transient depletion of macrophages altered local inflammatory response at the site of disc herniation.
Collapse
Affiliation(s)
- L Xiao
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA 22908, USA
| | - J Matharoo
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA 22908, USA
| | - J Chi
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA 22908, USA
| | - J Ma
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA 22908, USA
| | - M Chen
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA 22908, USA
| | - B Manley
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA 22908, USA
| | - P Xu
- Department of Pathology, University of Virginia, Charlottesville, VA 22908, USA
| | - W Shi
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA 22908, USA; Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22908, USA
| | - R A Felder
- Department of Pathology, University of Virginia, Charlottesville, VA 22908, USA
| | - S-S J Sung
- Department of Medicine and Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - L Jin
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA 22908, USA
| | - X Li
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA 22908, USA; Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22904, USA.
| |
Collapse
|
27
|
Mofarrah M, Jafari-Gharabaghlou D, Farhoudi-Sefidan-Jadid M, Zarghami N. Potential application of inorganic nano-materials in modulation of macrophage function: Possible application in bone tissue engineering. Heliyon 2023; 9:e16309. [PMID: 37292328 PMCID: PMC10245018 DOI: 10.1016/j.heliyon.2023.e16309] [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: 12/30/2022] [Revised: 05/11/2023] [Accepted: 05/12/2023] [Indexed: 06/10/2023] Open
Abstract
Nanomaterials indicate unique physicochemical properties for drug delivery in osteogenesis. Benefiting from high surface area grades, high volume ratio, ease of functionalization by biological targeting moieties, and small size empower nanomaterials to pass through biological barriers for efficient targeting. Inorganic nanomaterials for bone regeneration include inorganic synthetic polymers, ceramic nanoparticles, metallic nanoparticles, and magnetic nanoparticles. These nanoparticles can effectively modulate macrophage polarization and function, as one of the leading players in osteogenesis. Bone healing procedures in close cooperation with the immune system. Inflammation is one of the leading triggers of the bone fracture healing barrier. Macrophages commence anti-inflammatory signaling along with revascularization in the damaged site to promote the formation of a soft callus, bone mineralization, and bone remodeling. In this review, we will discuss the role of macrophages in bone hemostasis and regeneration. Furthermore, we will summarize the influence of the various inorganic nanoparticles on macrophage polarization and function in the benefit of osteogenesis.
Collapse
Affiliation(s)
- Mohsen Mofarrah
- Department of Medical Biotechnology, Faculty of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Davoud Jafari-Gharabaghlou
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdi Farhoudi-Sefidan-Jadid
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nosratollah Zarghami
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Biochemistry, Faculty of Medicine, Istanbul Aydin University, Istanbul, Turkey
| |
Collapse
|
28
|
Landini L, Marini M, Souza Monteiro de Araujo D, Romitelli A, Montini M, Albanese V, Titiz M, Innocenti A, Bianchini F, Geppetti P, Nassini R, De Logu F. Schwann Cell Insulin-like Growth Factor Receptor Type-1 Mediates Metastatic Bone Cancer Pain in Mice. Brain Behav Immun 2023; 110:348-364. [PMID: 36940752 DOI: 10.1016/j.bbi.2023.03.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/27/2023] [Accepted: 03/16/2023] [Indexed: 03/23/2023] Open
Abstract
Insulin growth factor-1 (IGF-1), an osteoclast-dependent osteolysis biomarker, contributes to metastatic bone cancer pain (MBCP), but the underlying mechanism is poorly understood. In mice, the femur metastasis caused by intramammary inoculation of breast cancer cells resulted in IGF-1 increase in femur and sciatic nerve, and IGF-1-dependent stimulus/non-stimulus-evoked pain-like behaviors. Adeno-associated virus-based shRNA selective silencing of IGF-1 receptor (IGF-1R) in Schwann cells, but not in dorsal root ganglion (DRG) neurons, attenuated pain-like behaviors. Intraplantar IGF-1 evoked acute nociception and mechanical/cold allodynia, which were reduced by selective IGF-1R silencing in DRG neurons and Schwann cells, respectively. Schwann cell IGF-1R signaling promoted an endothelial nitric oxide synthase-mediated transient receptor potential ankyrin 1 (TRPA1) activation and release of reactive oxygen species that, via macrophage-colony stimulating factor-dependent endoneurial macrophage expansion, sustained pain-like behaviors. Osteoclast derived IGF-1 initiates a Schwann cell-dependent neuroinflammatory response that sustains a proalgesic pathway that provides new options for MBCP treatment.
Collapse
Affiliation(s)
- Lorenzo Landini
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, University of Florence, Florence, 50139, Italy
| | - Matilde Marini
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, University of Florence, Florence, 50139, Italy
| | | | - Antonia Romitelli
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, University of Florence, Florence, 50139, Italy
| | - Marco Montini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Medical Genetics Unit, University of Florence, 50141, Florence, Italy
| | - Valentina Albanese
- Department of Environmental and Prevention Sciences - DEPS, University of Ferrara, Ferrara, 44121, Italy
| | - Mustafa Titiz
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, University of Florence, Florence, 50139, Italy
| | - Alessandro Innocenti
- Plastic and Reconstructive Microsurgery - Careggi University Hospital, Florence, 50139, Italy
| | - Francesca Bianchini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", Section of Experimental Pathology and Oncology, University of Florence, 50141, Florence, Italy
| | - Pierangelo Geppetti
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, University of Florence, Florence, 50139, Italy
| | - Romina Nassini
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, University of Florence, Florence, 50139, Italy.
| | - Francesco De Logu
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, University of Florence, Florence, 50139, Italy
| |
Collapse
|
29
|
Abstract
Tumour progression is modulated by the local microenvironment. This environment is populated by many immune cells, of which macrophages are among the most abundant. Clinical correlative data and a plethora of preclinical studies in mouse models of cancers have shown that tumour-associated macrophages (TAMs) play a cancer-promoting role. Within the primary tumour, TAMs promote tumour cell invasion and intravasation and tumour stem cell viability and induce angiogenesis. At the metastatic site, metastasis-associated macrophages promote extravasation, tumour cell survival and persistent growth, as well as maintain tumour cell dormancy in some contexts. In both the primary and metastatic sites, TAMs are suppressive to the activities of cytotoxic T and natural killer cells that have the potential to eradicate tumours. Such activities suggest that TAMs will be a major target for therapeutic intervention. In this Perspective article, we chronologically explore the evolution of our understanding of TAM biology put into the context of major enabling advances in macrophage biology.
Collapse
Affiliation(s)
| | - Jeffrey W Pollard
- MRC-Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
30
|
Gata6 + large peritoneal macrophages: an evolutionarily conserved sentinel and effector system for infection and injury. Trends Immunol 2023; 44:129-145. [PMID: 36623953 DOI: 10.1016/j.it.2022.12.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/05/2022] [Accepted: 12/06/2022] [Indexed: 01/08/2023]
Abstract
There are striking similarities between the sea urchin cavity macrophage-like phagocytes (coelomocytes) and mammalian cavity macrophages in not only their location, but also their behaviors. These cells are crucial for maintaining homeostasis within the cavity following a breach, filling the gap and functioning as a barrier between vital organs and the environment. In this review, we summarize the evolving literature regarding these Gata6+ large peritoneal macrophages (GLPMs), focusing on ontogeny, their responses to perturbations, including their rapid aggregation via coagulation, as well as scavenger receptor cysteine-rich domains and their potential roles in diseases, such as cancer. We challenge the 50-year old phenomenon of the 'macrophage disappearance reaction' (MDR) and propose the new term 'macrophage disturbance of homeostasis reaction' (MDHR), which may better describe this complex phenomenon.
Collapse
|
31
|
Lewis BE, Mulka A, Mao L, Sharafieh R, Qiao Y, Kesserwan S, Wu R, Kreutzer D, Klueh U. Insulin Derived Fibrils Induce Cytotoxicity in vitro and Trigger Inflammation in Murine Models. J Diabetes Sci Technol 2023; 17:163-171. [PMID: 34286629 PMCID: PMC9846386 DOI: 10.1177/19322968211033868] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Effective exogenous insulin delivery is the cornerstone of insulin dependent diabetes mellitus management. Recent literature indicates that commercial insulin-induced tissue reaction and cellular cytotoxicity may contribute to variability in blood glucose as well as permanent loss of injection or infusion site architecture and function. It is well accepted that insulin formulations are susceptible to mechanical and chemical stresses that lead to insulin fibril formation. This study aims to characterize in vitro and in vivo toxicity, as well as pro-inflammatory activity of insulin fibrils. METHOD In vitro cell culture evaluated cytotoxicity and fibril uptake by macrophages and our modified murine air-pouch model quantified inflammatory activity. The latter employed FLOW cytometry and histopathology to characterize fibril-induced inflammation in vivo, which included fibril uptake by inflammatory phagocytes. RESULTS These studies demonstrated that insulin derived fibrils are cytotoxic to cells in vitro. Furthermore, inflammation is induced in the murine air-pouch model in vivo and in response, macrophages uptake fibrils both in vitro and in vivo. CONCLUSIONS Administration of insulin fibrils can lead to cytotoxicity in macrophages. In vivo data demonstrate insulin fibrils to be pro-inflammatory which over time can lead to cumulative cell/tissue toxicity, inflammation, and destructive wound healing. Long term, these tissue reactions could contribute to loss of insulin injection site architecture and function.
Collapse
Affiliation(s)
- Brianne E. Lewis
- Department of Biomedical Engineering,
Integrative Biosciences Center. Wayne State University, Detroit, MI, USA
| | - Adam Mulka
- Department of Biomedical Engineering,
Integrative Biosciences Center. Wayne State University, Detroit, MI, USA
| | - Li Mao
- Department of Biomedical Engineering,
Integrative Biosciences Center. Wayne State University, Detroit, MI, USA
| | - Roshanak Sharafieh
- Department of Surgery, School of Medicine.
University of Connecticut, Farmington, CT, USA
| | - Yi Qiao
- Department of Surgery, School of Medicine.
University of Connecticut, Farmington, CT, USA
| | - Shereen Kesserwan
- Department of Biomedical Engineering,
Integrative Biosciences Center. Wayne State University, Detroit, MI, USA
| | - Rong Wu
- Department of Surgery, School of Medicine.
University of Connecticut, Farmington, CT, USA
| | - Don Kreutzer
- Department of Surgery, School of Medicine.
University of Connecticut, Farmington, CT, USA
| | - Ulrike Klueh
- Department of Biomedical Engineering,
Integrative Biosciences Center. Wayne State University, Detroit, MI, USA
- Ulrike Klueh, PhD, Department of Biomedical
Engineering, Integrative Biosciences Center, Wayne State University, 6135 Woodward Ave,
Detroit, MI 48202, USA.
| |
Collapse
|
32
|
Shen M, Kang Y. Cancer fitness genes: emerging therapeutic targets for metastasis. Trends Cancer 2023; 9:69-82. [PMID: 36184492 DOI: 10.1016/j.trecan.2022.08.007] [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: 07/07/2022] [Revised: 08/25/2022] [Accepted: 08/30/2022] [Indexed: 12/31/2022]
Abstract
Development of cancer therapeutics has traditionally focused on targeting driver oncogenes. Such an approach is limited by toxicity to normal tissues and treatment resistance. A class of 'cancer fitness genes' with crucial roles in metastasis have been identified. Elevated or altered activities of these genes do not directly cause cancer; instead, they relieve the stresses that tumor cells encounter and help them adapt to a changing microenvironment, thus facilitating tumor progression and metastasis. Importantly, as normal cells do not experience high levels of stress under physiological conditions, targeting cancer fitness genes is less likely to cause toxicity to noncancerous tissues. Here, we summarize the key features and function of cancer fitness genes and discuss their therapeutic potential.
Collapse
Affiliation(s)
- Minhong Shen
- Department of Pharmacology, Wayne State University School of Medicine, Michigan, MI, USA; Department of Oncology, Wayne State University School of Medicine and Tumor Biology and Microenvironment Research Program, Barbara Ann Karmanos Cancer Institute, Michigan, MI, USA.
| | - Yibin Kang
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA; Ludwig Institute for Cancer Research Princeton Branch, Princeton, NJ, USA.
| |
Collapse
|
33
|
Yu X, Ton AN, Niu Z, Morales BM, Chen J, Braz J, Lai MH, Barruet E, Liu H, Cheung K, Ali S, Chan T, Bigay K, Ho J, Nikolli I, Hansberry S, Wentworth K, Kriegstein A, Basbaum A, Hsiao EC. ACVR1-activating mutation causes neuropathic pain and sensory neuron hyperexcitability in humans. Pain 2023; 164:43-58. [PMID: 35442931 PMCID: PMC9582048 DOI: 10.1097/j.pain.0000000000002656] [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: 08/15/2021] [Revised: 03/01/2022] [Accepted: 04/08/2022] [Indexed: 01/09/2023]
Abstract
ABSTRACT Altered bone morphogenetic protein (BMP) signaling is associated with many musculoskeletal diseases. However, it remains unknown whether BMP dysfunction has direct contribution to debilitating pain reported in many of these disorders. Here, we identified a novel neuropathic pain phenotype in patients with fibrodysplasia ossificans progressiva (FOP), a rare autosomal-dominant musculoskeletal disorder characterized by progressive heterotopic ossification. Ninety-seven percent of these patients carry an R206H gain-of-function point mutation in the BMP type I receptor ACVR1 (ACVR1 R206H ), which causes neofunction to Activin A and constitutively activates signaling through phosphorylated SMAD1/5/8. Although patients with FOP can harbor pathological lesions in the peripheral and central nervous system, their etiology and clinical impact are unclear. Quantitative sensory testing of patients with FOP revealed significant heat and mechanical pain hypersensitivity. Although there was no major effect of ACVR1 R206H on differentiation and maturation of nociceptive sensory neurons (iSNs) derived from FOP induced pluripotent stem cells, both intracellular and extracellular electrophysiology analyses of the ACVR1 R206H iSNs displayed ACVR1-dependent hyperexcitability, a hallmark of neuropathic pain. Consistent with this phenotype, we recorded enhanced responses of ACVR1 R206H iSNs to TRPV1 and TRPA1 agonists. Thus, activated ACVR1 signaling can modulate pain processing in humans and may represent a potential target for pain management in FOP and related BMP pathway diseases.
Collapse
Affiliation(s)
- Xiaobing Yu
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, United States
| | - Amy N. Ton
- Division of Endocrinology and Metabolism, Department of Medicine, The Institute for Human Genetics, and the Program in Craniofacial Biology, University of California, San Francisco, CA, United States
| | - Zejun Niu
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, United States
- Department of Anesthesiology, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Blanca M. Morales
- Division of Endocrinology and Metabolism, Department of Medicine, The Institute for Human Genetics, and the Program in Craniofacial Biology, University of California, San Francisco, CA, United States
| | - Jiadong Chen
- Department of Neurology, University of California, San Francisco, CA, United States. Dr. Chen is now with the Department of Neurology of Second Affiliated Hospital, Centre for Neuroscience, Zhejiang University School of Medicine, Hangzhou, China
| | - Joao Braz
- Department of Anatomy, University of California San Francisco, San Francisco, CA, United States
| | - Michael H. Lai
- J. David Gladstone Institutes, San Francisco, CA, United States
| | - Emilie Barruet
- Division of Endocrinology and Metabolism, Department of Medicine, The Institute for Human Genetics, and the Program in Craniofacial Biology, University of California, San Francisco, CA, United States
| | - Hongju Liu
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, United States
- Department of Anesthesiology, Peking Union Medical College Hospital, Beijing, China
| | - Kin Cheung
- BioSAS Consulting, Inc, Wellesley, MA, United States
| | - Syed Ali
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, United States
| | - Tea Chan
- Division of Endocrinology and Metabolism, Department of Medicine, The Institute for Human Genetics, and the Program in Craniofacial Biology, University of California, San Francisco, CA, United States
| | - Katherine Bigay
- Division of Endocrinology and Metabolism, Department of Medicine, The Institute for Human Genetics, and the Program in Craniofacial Biology, University of California, San Francisco, CA, United States
| | - Jennifer Ho
- Division of Endocrinology and Metabolism, Department of Medicine, The Institute for Human Genetics, and the Program in Craniofacial Biology, University of California, San Francisco, CA, United States
| | - Ina Nikolli
- Division of Endocrinology and Metabolism, Department of Medicine, The Institute for Human Genetics, and the Program in Craniofacial Biology, University of California, San Francisco, CA, United States
| | - Steven Hansberry
- Division of Endocrinology and Metabolism, Department of Medicine, The Institute for Human Genetics, and the Program in Craniofacial Biology, University of California, San Francisco, CA, United States
- California Institute of Regenerative Medicine Bridges to Stem Cell Research Program, San Francisco State University, San Francisco, CA, United States
| | - Kelly Wentworth
- Division of Endocrinology and Metabolism, Department of Medicine, The Institute for Human Genetics, and the Program in Craniofacial Biology, University of California, San Francisco, CA, United States
| | - Arnold Kriegstein
- Department of Neurology, University of California, San Francisco, CA, United States. Dr. Chen is now with the Department of Neurology of Second Affiliated Hospital, Centre for Neuroscience, Zhejiang University School of Medicine, Hangzhou, China
| | - Allan Basbaum
- Department of Anatomy, University of California San Francisco, San Francisco, CA, United States
| | - Edward C. Hsiao
- Division of Endocrinology and Metabolism, Department of Medicine, The Institute for Human Genetics, and the Program in Craniofacial Biology, University of California, San Francisco, CA, United States
| |
Collapse
|
34
|
Gao X, Kady N, Wang C, Abdelrahman S, Gann P, Sverdlov M, Wolfe A, Brown N, Reneau J, Robida AM, Murga-Zamalloa C, Wilcox RA. Targeting Lymphoma-associated Macrophage Expansion via CSF1R/JAK Inhibition is a Therapeutic Vulnerability in Peripheral T-cell Lymphomas. CANCER RESEARCH COMMUNICATIONS 2022; 2:1727-1737. [PMID: 36970721 PMCID: PMC10035520 DOI: 10.1158/2767-9764.crc-22-0336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/25/2022] [Accepted: 12/08/2022] [Indexed: 12/16/2022]
Abstract
The reciprocal relationship between malignant T cells and lymphoma-associated macrophages (LAM) within the tumor microenvironment (TME) is unique, as LAMs are well poised to provide ligands for antigen, costimulatory, and cytokine receptors that promote T-cell lymphoma growth. Conversely, malignant T cells promote the functional polarization and homeostatic survival of LAM. Therefore, we sought to determine the extent to which LAMs are a therapeutic vulnerability in these lymphomas, and to identify effective therapeutic strategies for their depletion. We utilized complementary genetically engineered mouse models and primary peripheral T-cell lymphoma (PTCL) specimens to quantify LAM expansion and proliferation. A high-throughput screen was performed to identify targeted agents that effectively deplete LAM within the context of PTCL. We observed that LAMs are dominant constituents of the TME in PTCL. Furthermore, their dominance was explained, at least in part, by their proliferation and expansion in response to PTCL-derived cytokines. Importantly, LAMs are a true dependency in these lymphomas, as their depletion significantly impaired PTCL progression. These findings were extrapolated to a large cohort of human PTCL specimens where LAM proliferation was observed. A high-throughput screen demonstrated that PTCL-derived cytokines led to relative resistance to CSF1R selective inhibitors, and culminated in the identification of dual CSF1R/JAK inhibition as a novel therapeutic strategy to deplete LAM in these aggressive lymphomas. Malignant T cells promote the expansion and proliferation of LAM, which are a bone fide dependency in these lymphomas, and are effectively depleted with a dual CSF1R/JAK inhibitor. Significance LAMs are a therapeutic vulnerability, as their depletion impairs T-cell lymphoma disease progression. Pacritinib, a dual CSF1R/JAK inhibitor, effectively impaired LAM viability and expansion, prolonged survival in preclinical T-cell lymphoma models, and is currently being investigated as a novel therapeutic approach in these lymphomas.
Collapse
Affiliation(s)
- Xin Gao
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, Michigan
| | - Nermin Kady
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, Michigan
| | - Chenguang Wang
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, Michigan
| | - Suhaib Abdelrahman
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, Michigan
| | - Peter Gann
- Department of Pathology, University of Illinois Chicago, Chicago, Michigan
| | - Maria Sverdlov
- Department of Pathology, University of Illinois Chicago, Chicago, Michigan
| | - Ashley Wolfe
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, Michigan
| | - Noah Brown
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - John Reneau
- Department of Medicine, Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Aaron M. Robida
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan
| | | | - Ryan A. Wilcox
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
35
|
Shen Y, Zhang Y, Zhou Z, Wang J, Han D, Sun J, Chen G, Tang Q, Sun W, Chen L. Dysfunction of macrophages leads to diabetic bone regeneration deficiency. Front Immunol 2022; 13:990457. [PMID: 36311779 PMCID: PMC9613949 DOI: 10.3389/fimmu.2022.990457] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 10/03/2022] [Indexed: 11/22/2022] Open
Abstract
Insufficient bone matrix formation caused by diabetic chronic inflammation can result in bone nonunion, which is perceived as a worldwide epidemic, with a substantial socioeconomic and public health burden. Macrophages in microenvironment orchestrate the inflammation and launch the process of bone remodeling and repair, but aberrant activation of macrophages can drive drastic inflammatory responses during diabetic bone regeneration. In diabetes mellitus, the proliferation of resident macrophages in bone microenvironment is limited, while enhanced myeloid differentiation of hematopoietic stem cells (HSCs) leads to increased and constant monocyte recruitment and thus macrophages shift toward the classic pro-inflammatory phenotype, which leads to the deficiency of bone regeneration. In this review, we systematically summarized the anomalous origin of macrophages under diabetic conditions. Moreover, we evaluated the deficit of pro-regeneration macrophages in the diabetic inflammatory microenvironment. Finally, we further discussed the latest developments on strategies based on targeting macrophages to promote diabetic bone regeneration. Briefly, this review aimed to provide a basis for modulating the biological functions of macrophages to accelerate bone regeneration and rescue diabetic fracture healing in the future.
Collapse
Affiliation(s)
- Yufeng Shen
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillary Development and Regeneration, Wuhan, China
| | - Yifan Zhang
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillary Development and Regeneration, Wuhan, China
| | - Zheng Zhou
- Department of Stomatology, The First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Jinyu Wang
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillary Development and Regeneration, Wuhan, China
| | - Dong Han
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillary Development and Regeneration, Wuhan, China
| | - Jiwei Sun
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillary Development and Regeneration, Wuhan, China
| | - Guangjin Chen
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillary Development and Regeneration, Wuhan, China
| | - Qingming Tang
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillary Development and Regeneration, Wuhan, China
| | - Wei Sun
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillary Development and Regeneration, Wuhan, China
- *Correspondence: Lili Chen, ; Wei Sun,
| | - Lili Chen
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillary Development and Regeneration, Wuhan, China
- *Correspondence: Lili Chen, ; Wei Sun,
| |
Collapse
|
36
|
Krall JTW, Gibbs KW, Belfield L, Liu C, Purcell L, Bivona JJ, Poynter ME, Stapleton RD, Toth MJ, Files DC. Skeletal muscle macrophage ablation in mice. J Immunol Methods 2022; 509:113329. [PMID: 35933049 PMCID: PMC9748827 DOI: 10.1016/j.jim.2022.113329] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 07/01/2022] [Accepted: 08/01/2022] [Indexed: 10/16/2022]
Abstract
Macrophages are important mediators of skeletal muscle function in both healthy and diseased states. In vivo specific depletion of macrophages provides an experimental method to understand physiological and pathophysiological effects of macrophages. Systemic depletion of macrophages can deplete skeletal muscle macrophages but also alters systemic inflammatory responses and metabolism, which confounds the muscle specific effects of macrophage depletion. The primary aim of this manuscript is to evaluate two methods of murine intramuscular macrophage depletion in an acute lung injury-associated indirect skeletal muscle wasting mouse model. Adult C57BL/6 (WT) and Macrophage Fas-Induced Apoptosis (MaFIA, C57BL/6-Tg) mice received clodronate liposomes or the dimerization drug AP20187 through intramuscular injection of the tibialis anterior muscle compartment, respectively. Vehicle control was injected in the contralateral muscle. We demonstrate intramuscular AP20187 in the MaFIA mouse depletes macrophages but causes an infiltration of CD45 intermediate neutrophils. In contrast, intramuscular clodronate liposomes successfully depletes macrophages without an associated increase in CD45 intermediate cells. In conclusion, intramuscular clodronate is effective for selective depletion of muscle macrophages without eliciting acute inflammation seen with AP20187 in MaFIA mice. This technique is an important tool to study the functional roles of macrophages in skeletal muscle.
Collapse
Affiliation(s)
- Jennifer T W Krall
- Department of Internal Medicine, Section on Pulmonary, Critical Care, Allergy, and Immunologic Diseases, Wake Forest School of Medicine, Winston-Salem, NC, United States.
| | - Kevin W Gibbs
- Department of Internal Medicine, Section on Pulmonary, Critical Care, Allergy, and Immunologic Diseases, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Lanazha Belfield
- Department of Internal Medicine, Section on Pulmonary, Critical Care, Allergy, and Immunologic Diseases, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Chun Liu
- Department of Internal Medicine, Section on Pulmonary, Critical Care, Allergy, and Immunologic Diseases, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Lina Purcell
- Department of Internal Medicine, Section on Pulmonary, Critical Care, Allergy, and Immunologic Diseases, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Joseph J Bivona
- Department of Medicine, University of Vermont Larner College of Medicine, Burlington, VT, United States
| | - Matthew E Poynter
- Department of Medicine, University of Vermont Larner College of Medicine, Burlington, VT, United States
| | - Renee D Stapleton
- Department of Medicine, University of Vermont Larner College of Medicine, Burlington, VT, United States
| | - Michael J Toth
- Department of Medicine, University of Vermont Larner College of Medicine, Burlington, VT, United States; Department of Molecular Physiology and Biophysics, University of Vermont Larner College of Medicine, Burlington, VT, United States
| | - D Clark Files
- Department of Internal Medicine, Section on Pulmonary, Critical Care, Allergy, and Immunologic Diseases, Wake Forest School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
37
|
Tsai TL, Zhou TA, Hsieh YT, Wang JC, Cheng HK, Huang CH, Tsai PY, Fan HH, Feng HK, Huang YC, Lin CC, Lin CH, Lin CY, Dzhagalov IL, Hsu CL. Multiomics reveal the central role of pentose phosphate pathway in resident thymic macrophages to cope with efferocytosis-associated stress. Cell Rep 2022; 40:111065. [PMID: 35830797 DOI: 10.1016/j.celrep.2022.111065] [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: 12/23/2021] [Revised: 05/07/2022] [Accepted: 06/16/2022] [Indexed: 11/29/2022] Open
Abstract
Tissue-resident macrophages (TRMs) are heterogeneous cell populations found throughout the body. Depending on their location, they perform diverse functions maintaining tissue homeostasis and providing immune surveillance. To survive and function within, TRMs adapt metabolically to the distinct microenvironments. However, little is known about the metabolic signatures of TRMs. The thymus provides a nurturing milieu for developing thymocytes yet efficiently removes those that fail the selection, relying on the resident thymic macrophages (TMφs). This study harnesses multiomics analyses to characterize TMφs and unveils their metabolic features. We find that the pentose phosphate pathway (PPP) is preferentially activated in TMφs, responding to the reduction-oxidation demands associated with the efferocytosis of dying thymocytes. The blockade of PPP in Mφs leads to decreased efferocytosis, which can be rescued by reactive oxygen species (ROS) scavengers. Our study reveals the key role of the PPP in TMφs and underscores the importance of metabolic adaptation in supporting Mφ efferocytosis.
Collapse
Affiliation(s)
- Tsung-Lin Tsai
- Institute of Microbiology and Immunology, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan
| | - Tyng-An Zhou
- Institute of Microbiology and Immunology, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Yu-Ting Hsieh
- Institute of Microbiology and Immunology, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Ju-Chu Wang
- Institute of Microbiology and Immunology, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Hui-Kuei Cheng
- Institute of Microbiology and Immunology, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Chen-Hua Huang
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Pei-Yuan Tsai
- Institute of Microbiology and Immunology, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Hsiu-Han Fan
- Institute of Microbiology and Immunology, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Hsing-Kai Feng
- Institute of Microbiology and Immunology, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Yu-Chia Huang
- Institute of Microbiology and Immunology, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Chen-Ching Lin
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Chao-Hsiung Lin
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Chih-Yu Lin
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei 112, Taiwan
| | - Ivan L Dzhagalov
- Institute of Microbiology and Immunology, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Chia-Lin Hsu
- Institute of Microbiology and Immunology, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
38
|
Zhou X, Zhang Z, Jiang W, Hu M, Meng Y, Li W, Zhou X, Wang C. Naringenin is a Potential Anabolic Treatment for Bone Loss by Modulating Osteogenesis, Osteoclastogenesis, and Macrophage Polarization. Front Pharmacol 2022; 13:872188. [PMID: 35586056 PMCID: PMC9108355 DOI: 10.3389/fphar.2022.872188] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 03/31/2022] [Indexed: 11/13/2022] Open
Abstract
Bone undergoes constant remodeling of formation by osteoblasts and resorption by osteoclasts. In particular, macrophages have been reported to play an essential role in the regulation of bone homeostasis and regeneration. Naringenin, the predominant flavanone in citrus fruits, is reported to exert anti-inflammatory, anti-osteoclastic, and osteogenic effects. However, whether naringenin could modulate the crosstalk between macrophages and osteoblasts/osteoclasts remains to be investigated. In this study, we confirmed that naringenin enhanced osteogenesis and inhibited osteoclastogenesis directly. Naringenin promoted M2 transition and the secretion of osteogenic cytokines including IL-4, IL-10, BMP2, and TGF-β, while suppressing LPS-induced M1 polarization and the production of proinflammatory factors such as TNF-α and IL-1β. In addition, the coculture of primary bone mesenchymal stem cells (BMSCs)/bone marrow monocytes (BMMs) with macrophages showed that the naringenin-treated medium significantly enhanced osteogenic differentiation and impeded osteoclastic differentiation in both inflammatory and non-inflammatory environment. Moreover, in vivo experiments demonstrated that naringenin remarkably reversed LPS-induced bone loss and assisted the healing of calvarial defect. Taken together, naringenin serves as a potential anabolic treatment for pathological bone loss.
Collapse
Affiliation(s)
- Xin Zhou
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Zheng Zhang
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
- College of Basic Medicine, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Weiwei Jiang
- Department of Critical Care Medicine, Changzheng Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Miao Hu
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
- College of Basic Medicine, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Yichen Meng
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Wenfang Li
- Department of Critical Care Medicine, Changzheng Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
- *Correspondence: Wenfang Li, ; Xuhui Zhou, ; Ce Wang,
| | - Xuhui Zhou
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
- *Correspondence: Wenfang Li, ; Xuhui Zhou, ; Ce Wang,
| | - Ce Wang
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
- *Correspondence: Wenfang Li, ; Xuhui Zhou, ; Ce Wang,
| |
Collapse
|
39
|
Saleh LS, Amer LD, Thompson BJ, Danhorn T, Knapp JR, Gibbings SL, Thomas S, Barthel L, O'Connor BP, Janssen WJ, Alper S, Bryant SJ. Mapping Macrophage Polarization and Origin during the Progression of the Foreign Body Response to a Poly(ethylene glycol) Hydrogel Implant. Adv Healthc Mater 2022; 11:e2102209. [PMID: 34967497 PMCID: PMC9081184 DOI: 10.1002/adhm.202102209] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 12/04/2021] [Indexed: 11/10/2022]
Abstract
Poly(ethylene glycol) (PEG) hydrogels hold promise for in vivo applications but induce a foreign body response (FBR). While macrophages are key in the FBR, many questions remain. This study investigates temporal changes in the transcriptome of implant-associated monocytes and macrophages. Proinflammatory pathways are upregulated in monocytes compared to control monocytes but subside by day 28. Macrophages are initially proinflammatory but shift to a profibrotic state by day 14, coinciding with fibrous capsule emergence. Next, this study assesses the origin of macrophages responsible for fibrous encapsulation using wildtype, C-C Motif Chemokine Receptor 2 (CCR2)-/- mice that lack recruited macrophages, and Macrophage Fas-Induced Apoptosis (MaFIA) mice that enable macrophage ablation. Subpopulations of recruited and tissue-resident macrophages are identified. Fibrous encapsulation proceeds in CCR2-/- mice similar to wildtype mice. However, studies in MaFIA mice indicate that macrophages are necessary for fibrous capsule formation. These findings suggest that macrophage origin impacts the FBR progression and provides evidence that tissue-resident macrophages and not the recruited macrophages may drive fibrosis in the FBR to PEG hydrogels. This study demonstrates that implant-associated monocytes and macrophages have temporally distinct transcriptomes in the FBR and that profibrotic pathways associated with macrophages may be enriched in tissue-resident macrophages.
Collapse
Affiliation(s)
- Leila S. Saleh
- Department of Chemical and Biological Engineering University of Colorado at Boulder 3415 Colorado Ave Boulder CO 80309 USA
| | - Luke D. Amer
- Department of Chemical and Biological Engineering University of Colorado at Boulder 3415 Colorado Ave Boulder CO 80309 USA
| | - Brittany J. Thompson
- Materials Science and Engineering Program University of Colorado at Boulder 3415 Colorado Ave Boulder CO 80309 USA
| | - Thomas Danhorn
- Center for Genes Environment and Health National Jewish Health 1400 Jackson St Denver CO 80206 USA
| | - Jennifer R. Knapp
- Center for Genes Environment and Health National Jewish Health 1400 Jackson St Denver CO 80206 USA
| | | | - Stacey Thomas
- Division of Pulmonary Sciences and Critical Care Medicine University of Colorado Denver Aurora CO 80045 USA
| | - Lea Barthel
- Division of Pulmonary Sciences and Critical Care Medicine University of Colorado Denver Aurora CO 80045 USA
| | - Brian P. O'Connor
- Center for Genes Environment and Health National Jewish Health 1400 Jackson St Denver CO 80206 USA
- Department of Immunology and Genomic Medicine National Jewish Health Denver CO 80206 USA
| | - William J. Janssen
- Division of Pulmonary Sciences and Critical Care Medicine University of Colorado Denver Aurora CO 80045 USA
- Division of Pulmonary Sleep, and Critical Care Medicine National Jewish Health Denver CO 80206 USA
| | - Scott Alper
- Center for Genes Environment and Health National Jewish Health 1400 Jackson St Denver CO 80206 USA
- Department of Immunology and Genomic Medicine National Jewish Health Denver CO 80206 USA
- Department of Immunology and Microbiology University of Colorado School of Medicine Aurora CO 80045 USA
| | - Stephanie J. Bryant
- Department of Chemical and Biological Engineering University of Colorado at Boulder 3415 Colorado Ave Boulder CO 80309 USA
- Materials Science and Engineering Program University of Colorado at Boulder 3415 Colorado Ave Boulder CO 80309 USA
- BioFrontiers Institute University of Colorado at Boulder Boulder CO 80309 USA
| |
Collapse
|
40
|
Burrack AL, Schmiechen ZC, Patterson MT, Miller EA, Spartz EJ, Rollins MR, Raynor JF, Mitchell JS, Kaisho T, Fife BT, Stromnes IM. Distinct myeloid antigen-presenting cells dictate differential fates of tumor-specific CD8+ T cells in pancreatic cancer. JCI Insight 2022; 7:e151593. [PMID: 35393950 PMCID: PMC9057584 DOI: 10.1172/jci.insight.151593] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 02/18/2022] [Indexed: 01/12/2023] Open
Abstract
We investigate how myeloid subsets differentially shape immunity to pancreatic ductal adenocarcinoma (PDA). We show that tumor antigenicity sculpts myeloid cell composition and functionality. Antigenicity promotes accumulation of type 1 dendritic cells (cDC1), which is driven by Xcr1 signaling, and overcomes macrophage-mediated suppression. The therapeutic activity of adoptive T cell therapy or programmed cell death ligand 1 blockade required cDC1s, which sustained splenic Klrg1+ cytotoxic antitumor T cells and functional intratumoral T cells. KLRG1 and cDC1 genes correlated in human tumors, and PDA patients with high intratumoral KLRG1 survived longer than patients with low intratumoral KLRG1. The immunotherapy CD40 agonist also required host cDC1s for maximal therapeutic benefit. However, CD40 agonist exhibited partial therapeutic benefit in cDC1-deficient hosts and resulted in priming of tumor-specific yet atypical CD8+ T cells with a regulatory phenotype and that failed to participate in tumor control. Monocyte/macrophage depletion using clodronate liposomes abrogated T cell priming yet enhanced the antitumor activity of CD40 agonist in cDC1-deficient hosts via engagement of innate immunity. In sum, our study supports that cDC1s are essential for sustaining effective antitumor T cells and supports differential roles for cDC1s and monocytes/macrophages in instructing T cell fate and immunotherapy response.
Collapse
Affiliation(s)
- Adam L. Burrack
- Department of Microbiology and Immunology
- Center for Immunology
| | | | | | - Ebony A. Miller
- Department of Microbiology and Immunology
- Center for Immunology
| | | | | | | | - Jason S. Mitchell
- Center for Immunology
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Tsuneyasu Kaisho
- Department of Immunology, Institute of Advanced Medicine, Wakayama Medical University, Kimiidera, Wakayama, Japan
| | - Brian T. Fife
- Center for Immunology
- Department of Medicine, and
- Masonic Cancer Center, and
| | - Ingunn M. Stromnes
- Department of Microbiology and Immunology
- Center for Immunology
- Masonic Cancer Center, and
- Center for Genome Engineering, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| |
Collapse
|
41
|
Chen M, Tian X, Xu L, Wu R, He H, Zhu H, Xu W, Wei CJ. Membrane tethering of CreER decreases uninduced cell labeling and cytotoxicity while maintaining recombination efficiency. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 27:1078-1091. [PMID: 35228901 PMCID: PMC8851158 DOI: 10.1016/j.omtn.2022.01.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 01/28/2022] [Indexed: 02/05/2023]
Abstract
Genetic lineage tracing is indispensable to unraveling the origin, fate, and plasticity of cells. However, the intrinsic leakiness in the CreER-loxP system raises concerns on data interpretation. Here, we reported the generation of a novel dual inducible CreER-loxP system with superior labeling characteristics. This two-component system consists of membrane localized CreER (mCreER: CD8α-FRB-CS-CreER) and TEV protease (mTEVp: CD8α-FKBP-TEVp), which are fusion proteins incorporated with the chemically induced dimerization machinery. Rapamycin and tamoxifen induce sequential dimerization of FKBP and FRB, cleavage of CreER from the membrane, and translocation into the nucleus. The labeling leakiness in Ad293 cells reduced dramatically from more than 70% to less than 5%. This tight labeling feature depends largely on the association of mCreER with HSP90, which conceals the TEV protease cutting site between FRB and CreER and thus preventing uninduced cleavage of the membrane-tethering CreER. Membrane-bound CreER also diminished significantly cytotoxicity. Our studies showed mCreER under the control of the rat insulin promoter increased labeling specificity in MIN6 islet beta-cells. Viability and insulin secretion of MIN6 cells remained intact. Our results demonstrate that this novel system can provide more stringent temporal and spatial control of gene expression and will be useful in cell fate probing.
Collapse
Affiliation(s)
- Mianqiao Chen
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou, Guangdong 515063, China
| | - Xiong Tian
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou, Guangdong 515063, China
| | - Liqun Xu
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou, Guangdong 515063, China
| | - Ruolan Wu
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou, Guangdong 515063, China
| | - Haoming He
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou, Guangdong 515063, China
| | - Haibao Zhu
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou, Guangdong 515063, China
| | - Wencan Xu
- Department of Endocrinology, the First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Chi-ju Wei
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou, Guangdong 515063, China
- Multidisciplinary Research Center, Shantou University, Shantou, Guangdong 515063, China
- Corresponding author Chi-ju Wei, PhD, Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou, Guangdong 515063, China.
| |
Collapse
|
42
|
Disruption of Hyaluronic Acid in Skeletal Muscle Induces Decreased Voluntary Activity via Chemosensitive Muscle Afferent Sensitization in Male Mice. eNeuro 2022; 9:ENEURO.0522-21.2022. [PMID: 35387844 PMCID: PMC9014980 DOI: 10.1523/eneuro.0522-21.2022] [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: 11/16/2021] [Revised: 02/25/2022] [Accepted: 03/09/2022] [Indexed: 11/21/2022] Open
Abstract
PEGPH20, a human recombinant hyaluronidase, has been proposed as a coadjutant to pancreatic cancer chemotherapy. In early trials, patients reported increased widespread muscle pain as the main adverse reaction to PEGPH20. To understand how PEGPH20 caused musculoskeletal pain, we systemically administered PEGPH20 to male mice and measured voluntary wheel activity and pain-related behaviors. These were paired with ex vivo electrophysiology of primary sensory neurons, whole DRG real-time PCR, and immunohistochemistry of hindpaw muscle. PEGPH20 induced significantly lower wheel running, compared with vehicle-treated animals, and decreased mechanical withdrawal thresholds 5 d after PEGPH20 injections. Chemo-sensory muscle afferents showed increased responses to noxious chemical stimulation of their receptive fields (RFs) in the PEGPH20-treated group. This was correlated with upregulation of the NGF receptor TrkA, the transient receptor potential vanilloid type 1 (TRPV1) channel and ATP-sensitive channel P2X3 in the DRG. Immunohistochemistry of hindpaw muscles revealed damage to the muscle architecture and extensive infiltration of the tissue by cells of the myelomonocytic lineage 3 d after PEGPH20 injection. Peripheral macrophage ablation in macrophage Fas-induced apoptosis (MaFIA) mice, however, did not prevent the decreased voluntary activity and instead caused even lower levels of running. These results suggest that disruption of hyaluronic acid (HA) within the muscle extracellular matrix (ECM) sensitizes chemo-nociceptive muscle afferents possibly leading to altered pain-like behaviors. Ablation experiments suggest macrophages are necessary for adequate recovery of voluntary activity after HA disruption. These data support a role for HA and macrophages in tissue integrity and muscle pain development in patients taking PEGPH20.
Collapse
|
43
|
Zhou TA, Hsu HP, Tu YH, Cheng HK, Lin CY, Chen NJ, Tsai JW, Robey EA, Huang HC, Hsu CL, Dzhagalov IL. Thymic macrophages consist of two populations with distinct localization and origin. eLife 2022; 11:75148. [PMID: 36449334 PMCID: PMC9754631 DOI: 10.7554/elife.75148] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 11/29/2022] [Indexed: 12/03/2022] Open
Abstract
Tissue-resident macrophages are essential to protect from pathogen invasion and maintain organ homeostasis. The ability of thymic macrophages to engulf apoptotic thymocytes is well appreciated, but little is known about their ontogeny, maintenance, and diversity. Here, we characterized the surface phenotype and transcriptional profile of these cells and defined their expression signature. Thymic macrophages were most closely related to spleen red pulp macrophages and Kupffer cells and shared the expression of the transcription factor (TF) SpiC with these cells. Single-cell RNA sequencing (scRNA-Seq) showed that the macrophages in the adult thymus are composed of two populations distinguished by the expression of Timd4 and Cx3cr1. Remarkably, Timd4+ cells were located in the cortex, while Cx3cr1+ macrophages were restricted to the medulla and the cortico-medullary junction. Using shield chimeras, transplantation of embryonic thymuses, and genetic fate mapping, we found that the two populations have distinct origins. Timd4+ thymic macrophages are of embryonic origin, while Cx3cr1+ macrophages are derived from adult hematopoietic stem cells. Aging has a profound effect on the macrophages in the thymus. Timd4+ cells underwent gradual attrition, while Cx3cr1+ cells slowly accumulated with age and, in older mice, were the dominant macrophage population in the thymus. Altogether, our work defines the phenotype, origin, and diversity of thymic macrophages.
Collapse
Affiliation(s)
- Tyng-An Zhou
- Institute of Microbiology and Immunology, National Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Hsuan-Po Hsu
- Institute of Microbiology and Immunology, National Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Yueh-Hua Tu
- Bioinformatics Program, Taiwan International Graduate Program, Institute of Information Science, Academia SinicaTaipeiTaiwan,Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan UniversityTaipeiTaiwan
| | - Hui-Kuei Cheng
- Institute of Microbiology and Immunology, National Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Chih-Yu Lin
- Institute of Microbiology and Immunology, National Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Nien-Jung Chen
- Institute of Microbiology and Immunology, National Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Jin-Wu Tsai
- Brain Research Center, National Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Ellen A Robey
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | - Hsuan-Cheng Huang
- Bioinformatics Program, Taiwan International Graduate Program, Institute of Information Science, Academia SinicaTaipeiTaiwan,Institute of Biomedical Informatics, National Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Chia-Lin Hsu
- Institute of Microbiology and Immunology, National Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Ivan L Dzhagalov
- Institute of Microbiology and Immunology, National Yang Ming Chiao Tung UniversityTaipeiTaiwan
| |
Collapse
|
44
|
Ehnert S, Relja B, Schmidt-Bleek K, Fischer V, Ignatius A, Linnemann C, Rinderknecht H, Huber-Lang M, Kalbitz M, Histing T, Nussler AK. Effects of immune cells on mesenchymal stem cells during fracture healing. World J Stem Cells 2021; 13:1667-1695. [PMID: 34909117 PMCID: PMC8641016 DOI: 10.4252/wjsc.v13.i11.1667] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/31/2021] [Accepted: 09/03/2021] [Indexed: 02/06/2023] Open
Abstract
In vertebrates, bone is considered an osteoimmune system which encompasses functions of a locomotive organ, a mineral reservoir, a hormonal organ, a stem cell pool and a cradle for immune cells. This osteoimmune system is based on cooperatively acting bone and immune cells, cohabitating within the bone marrow. They are highly interdependent, a fact that is confounded by shared progenitors, mediators, and signaling pathways. Successful fracture healing requires the participation of all the precursors, immune and bone cells found in the osteoimmune system. Recent evidence demonstrated that changes of the immune cell composition and function may negatively influence bone healing. In this review, first the interplay between different immune cell types and osteoprogenitor cells will be elaborated more closely. The separate paragraphs focus on the specific cell types, starting with the cells of the innate immune response followed by cells of the adaptive immune response, and the complement system as mediator between them. Finally, a brief overview on the challenges of preclinical testing of immune-based therapeutic strategies to support fracture healing will be given.
Collapse
Affiliation(s)
- Sabrina Ehnert
- Siegfried Weller Research Institute at the BG Trauma Center Tübingen, Department of Trauma and Reconstructive Surgery, University of Tübingen, Tübingen 72076, Germany
| | - Borna Relja
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto-von-Guericke University, Magdeburg 39120, Germany
| | - Katharina Schmidt-Bleek
- Julius Wolff Institute and Berlin Institute of Health Center of Regenerative Therapies, Charité - University Medicine Berlin, Berlin 13353, Germany
| | - Verena Fischer
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm 89091, Germany
| | - Anita Ignatius
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, Ulm 89091, Germany
| | - Caren Linnemann
- Siegfried Weller Research Institute at the BG Trauma Center Tübingen, Department of Trauma and Reconstructive Surgery, University of Tübingen, Tübingen 72076, Germany
| | - Helen Rinderknecht
- Siegfried Weller Research Institute at the BG Trauma Center Tübingen, Department of Trauma and Reconstructive Surgery, University of Tübingen, Tübingen 72076, Germany
| | - Markus Huber-Lang
- Institute for Clinical and Experimental Trauma-Immunology (ITI), University Hospital Ulm, Ulm 89091, Germany
| | - Miriam Kalbitz
- Department of Trauma and Orthopedic Surgery, University Hospital Erlangen Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen 91054, Germany
| | - Tina Histing
- Siegfried Weller Research Institute at the BG Trauma Center Tübingen, Department of Trauma and Reconstructive Surgery, University of Tübingen, Tübingen 72076, Germany
| | - Andreas K Nussler
- Siegfried Weller Research Institute at the BG Trauma Center Tübingen, Department of Trauma and Reconstructive Surgery, University of Tübingen, Tübingen 72076, Germany
| |
Collapse
|
45
|
Schlundt C, Fischer H, Bucher CH, Rendenbach C, Duda GN, Schmidt-Bleek K. The multifaceted roles of macrophages in bone regeneration: A story of polarization, activation and time. Acta Biomater 2021; 133:46-57. [PMID: 33974949 DOI: 10.1016/j.actbio.2021.04.052] [Citation(s) in RCA: 130] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 03/26/2021] [Accepted: 04/27/2021] [Indexed: 12/12/2022]
Abstract
To present knowledge, macrophages are found in all tissues of the human body. They are a cell population with high plasticity which come with a multitude of functions which appear to be adapted to the respective tissue niche and micro-environment in which they reside. Bone harbors multiple macrophage subpopulations, with the osteoclasts as classical representative of a bone resorbing cells and osteomacs as a bone tissue resident macrophage first described by the expression of F4/80. Both subtypes are found throughout all phases in bone healing. In vivo data on bone regeneration have demonstrated their essential role in initiating the healing cascade (inflammatory phase) but also of the later phases of healing (e.g. endochondral and intramembranous bone formation). To participate in such diverse processes macrophages have to be highly plastic in their functionality. Thus, the widely used M1/M2 paradigm to distinguish macrophage subpopulations may not mirror the comprehensive role of the dynamics of macrophage plasticity. From a clinical perspective it is especially relevant to distinguish what drives macrophages in impaired healing scenarios, implant loosening or infections, where their specific role of a misbalanced inflammatory setting is so far only partially known. With this review we aim at illustrating current knowledge and gaps of knowledge on macrophage plasticity and function during the cascades of regeneration and reconstitution of bone tissue. We propose aspects of the known biological mechanisms of macrophages and their specific subsets that might serve as targets to control their function in impaired healing and eventually support a scar-free regeneration. STATEMENT OF SIGNIFICANCE: Macrophages are essential for successful regeneration. In scar-free healing such as in bone, a complete failure of healing was shown if macrophages were depleted; the M1/M2 switch appears to be key to the progression from pro-inflammation to regeneration. However, experimental data illustrate that the classical M1/M2 paradigm does not completely mirror the complexity of observed macrophage functions during bone healing and thus demands a broader perspective. Within this review we discuss the high degree of plasticity of macrophages and the relevant contribution of the different and more specific M2 subtypes (M2a-M2f) during (bone) regeneration. It summarizes the versatile roles of macrophages in skeletal regeneration and thereby highlights potential target points for immunomodulatory approaches to enable or even foster bone repair.
Collapse
|
46
|
Whitaker R, Hernaez-Estrada B, Hernandez RM, Santos-Vizcaino E, Spiller KL. Immunomodulatory Biomaterials for Tissue Repair. Chem Rev 2021; 121:11305-11335. [PMID: 34415742 DOI: 10.1021/acs.chemrev.0c00895] [Citation(s) in RCA: 109] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
All implanted biomaterials are targets of the host's immune system. While the host inflammatory response was once considered a detrimental force to be blunted or avoided, in recent years, it has become a powerful force to be leveraged to augment biomaterial-tissue integration and tissue repair. In this review, we will discuss the major immune cells that mediate the inflammatory response to biomaterials, with a focus on how biomaterials can be designed to modulate immune cell behavior to promote biomaterial-tissue integration. In particular, the intentional activation of monocytes and macrophages with controlled timing, and modulation of their interactions with other cell types involved in wound healing, have emerged as key strategies to improve biomaterial efficacy. To this end, careful design of biomaterial structure and controlled release of immunomodulators can be employed to manipulate macrophage phenotype for the maximization of the wound healing response with enhanced tissue integration and repair, as opposed to a typical foreign body response characterized by fibrous encapsulation and implant isolation. We discuss current challenges in the clinical translation of immunomodulatory biomaterials, such as limitations in the use of in vitro studies and animal models to model the human immune response. Finally, we describe future directions and opportunities for understanding and controlling the biomaterial-immune system interface, including the application of new imaging tools, new animal models, the discovery of new cellular targets, and novel techniques for in situ immune cell reprogramming.
Collapse
Affiliation(s)
- Ricardo Whitaker
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, Pennsylvania 19104, United States
| | - Beatriz Hernaez-Estrada
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, Pennsylvania 19104, United States.,NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz 01006, Spain
| | - Rosa Maria Hernandez
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz 01006, Spain.,Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz 01006, Spain
| | - Edorta Santos-Vizcaino
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz 01006, Spain.,Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz 01006, Spain
| | - Kara L Spiller
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
47
|
Kuo PJ, Rau CS, Wu SC, Lin CW, Huang LH, Lu TH, Wu YC, Wu CJ, Tsai CW, Hsieh CH. Exosomes Secreted by Adipose-Derived Stem Cells Following FK506 Stimulation Reduce Autophagy of Macrophages in Spine after Nerve Crush Injury. Int J Mol Sci 2021; 22:9628. [PMID: 34502537 PMCID: PMC8431814 DOI: 10.3390/ijms22179628] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/02/2021] [Accepted: 09/03/2021] [Indexed: 12/16/2022] Open
Abstract
Macrophages emerge in the milieu around innervated neurons after nerve injuries. Following nerve injury, autophagy is induced in macrophages and affects the regulation of inflammatory responses. It is closely linked to neuroinflammation, while the immunosuppressive drug tacrolimus (FK506) enhances nerve regeneration following nerve crush injury and nerve allotransplantation with additional neuroprotective and neurotrophic functions. The combined use of FK506 and adipose-derived stem cells (ADSCs) was employed in cell therapy for organ transplantation and vascularized composite allotransplantation. This study aimed to investigate the topical application of exosomes secreted by ADSCs following FK506 treatment (ADSC-F-exo) to the injured nerve in a mouse model of sciatic nerve crush injury. Furthermore, isobaric tags for relative and absolute quantitation (iTRAQ) were used to profile the potential exosomal proteins involved in autophagy. Immunohistochemical analysis revealed that nerve crush injuries significantly induced autophagy in the dorsal root ganglia and dorsal horn of the spinal segments. Locally applied ADSC-F-exo significantly reduced autophagy of macrophages in the spinal segments after nerve crush injury. Proteomic analysis showed that of the 22 abundant exosomal proteins detected in ADSC-F-exo, heat shock protein family A member 8 (HSPA8) and eukaryotic translation elongation factor 1 alpha 1 (EEF1A1) are involved in exosome-mediated autophagy reduction.
Collapse
Affiliation(s)
- Pao-Jen Kuo
- Department of Plastic Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan; (P.-J.K.); (C.-W.L.); (T.-H.L.); (Y.-C.W.); (C.-J.W.); (C.-W.T.)
| | - Cheng-Shyuan Rau
- Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan; (C.-S.R.); (L.-H.H.)
| | - Shao-Chun Wu
- Department of Anesthesiology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan;
| | - Chia-Wei Lin
- Department of Plastic Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan; (P.-J.K.); (C.-W.L.); (T.-H.L.); (Y.-C.W.); (C.-J.W.); (C.-W.T.)
| | - Lien-Hung Huang
- Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan; (C.-S.R.); (L.-H.H.)
| | - Tsu-Hsiang Lu
- Department of Plastic Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan; (P.-J.K.); (C.-W.L.); (T.-H.L.); (Y.-C.W.); (C.-J.W.); (C.-W.T.)
| | - Yi-Chan Wu
- Department of Plastic Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan; (P.-J.K.); (C.-W.L.); (T.-H.L.); (Y.-C.W.); (C.-J.W.); (C.-W.T.)
| | - Chia-Jung Wu
- Department of Plastic Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan; (P.-J.K.); (C.-W.L.); (T.-H.L.); (Y.-C.W.); (C.-J.W.); (C.-W.T.)
| | - Chia-Wen Tsai
- Department of Plastic Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan; (P.-J.K.); (C.-W.L.); (T.-H.L.); (Y.-C.W.); (C.-J.W.); (C.-W.T.)
| | - Ching-Hua Hsieh
- Department of Plastic Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan; (P.-J.K.); (C.-W.L.); (T.-H.L.); (Y.-C.W.); (C.-J.W.); (C.-W.T.)
- Center for Vascularized Composite Allotransplantation, Chang Gung Memorial Hospital, LinKou 33333, Taiwan
| |
Collapse
|
48
|
Yamashita M, Niisato M, Kawasaki Y, Karaman S, Robciuc MR, Shibata Y, Ishida Y, Nishio R, Masuda T, Sugai T, Ono M, Tuder RM, Alitalo K, Yamauchi K. VEGF-C/VEGFR-3 signaling in macrophages ameliorates acute lung injury. Eur Respir J 2021; 59:13993003.00880-2021. [PMID: 34446463 DOI: 10.1183/13993003.00880-2021] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 08/14/2021] [Indexed: 11/05/2022]
Abstract
RATIONALE Successful recovery from acute lung injury requires inhibition of neutrophil influx and clearance of apoptotic neutrophils. However, the mechanisms underlying recovery remain unclear. OBJECTIVES We investigated the ameliorative effects of vascular endothelial growth factor receptor-3 (VEGFR-3)/VEGF-C signaling in macrophages in lipopolysaccharide-induced lung injury. METHODS Lipopolysaccharides were intranasally injected into wild-type and transgenic mice. Gain- and loss- of VEGF-C/VEGFR-3 signaling function experiments employed adenovirus-mediated intranasal delivery of VEGF-C (Ad-VEGF-C vector) and soluble VEGFR-3, or, anti-VEGFR-3 blocking antibodies and mice with a deletion of VEGFR-3 in myeloid cells. MEASUREMENTS AND MAIN RESULTS The early phase of lung injury was significantly alleviated by the overexpression of VEGF-C with increased levels of bronchoalveolar lavage fluid (BALF) interleukin (IL)-10, but worsened in the later phase by VEGFR-3 inhibition upon administration of Ad-sVEGFR-3 vector. Injection of anti-VEGFR-3 antibodies to the mice in the resolution phase inhibited recovery from lung injury. The VEGFR-3 deleted mice had a shorter survival time than littermates and more severe lung injury in the resolution phase. Alveolar macrophages in the resolution phase digested most of extrinsic apoptotic neutrophils, and VEGF-C/VEGFR-3 signaling increased efferocytosis via upregulation of integrin alpha v in the macrophages. We also found that incubation with BALF from acute respiratory distress syndrome (ARDS) patients, but not from controls, decreases VEGFR-3 expression and the efficiency of IL-10 expression and efferocytosis in human monocyte-derived macrophages. CONCLUSIONS VEGFR-3/VEGF-C signaling in macrophages ameliorates experimental lung injury. This mechanism may provide an explanation also for ARDS resolution.
Collapse
Affiliation(s)
- Masahiro Yamashita
- Department of Pulmonary Medicine, Allergy and Immunological Diseases, Iwate Medical University School of Medicine, Morioka, Japan
| | - Miyuki Niisato
- Department of Pulmonary Medicine, Allergy and Immunological Diseases, Iwate Medical University School of Medicine, Morioka, Japan
| | - Yasushi Kawasaki
- Department of Health Chemistry, Iwate Medical University School of Pharmacology, Shiwa, Japan
| | - Sinem Karaman
- Wihuri Research Institute and Translational Cancer Medicine Program, University of Helsinki, Finland
| | - Marius R Robciuc
- Wihuri Research Institute and Translational Cancer Medicine Program, University of Helsinki, Finland
| | - Yuji Shibata
- Department of Pathology, Iwate Medical University School of Medicine, Japan
| | - Yoji Ishida
- Department of Hematology, Iwate Medical University School of Medicine, Japan
| | | | - Tomoyuki Masuda
- Department of Pathology, Iwate Medical University School of Medicine, Japan
| | - Tamotsu Sugai
- Department of Pathology, Iwate Medical University School of Medicine, Japan
| | - Masao Ono
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Rubin M Tuder
- Program in Translational Lung Research, Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado School of Medicine, CO, USA
| | - Kari Alitalo
- Wihuri Research Institute and Translational Cancer Medicine Program, University of Helsinki, Finland
| | - Kohei Yamauchi
- Department of Pulmonary Medicine, Allergy and Immunological Diseases, Iwate Medical University School of Medicine, Morioka, Japan
| |
Collapse
|
49
|
Liu X, Shi GP, Guo J. Innate Immune Cells in Pressure Overload-Induced Cardiac Hypertrophy and Remodeling. Front Cell Dev Biol 2021; 9:659666. [PMID: 34368120 PMCID: PMC8343105 DOI: 10.3389/fcell.2021.659666] [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: 01/28/2021] [Accepted: 06/28/2021] [Indexed: 12/23/2022] Open
Abstract
Pressure overload and heart failure are among the leading causes of cardiovascular morbidity and mortality. Accumulating evidence suggests that inflammatory cell activation and release of inflammatory mediators are of vital importance during the pathogenesis of these cardiac diseases. Yet, the roles of innate immune cells and subsequent inflammatory events in these processes remain poorly understood. Here, we outline the possible underlying mechanisms of innate immune cell participation, including mast cells, macrophages, monocytes, neutrophils, dendritic cells, eosinophils, and natural killer T cells in these pathological processes. Although these cells accumulate in the atrium or ventricles at different time points after pressure overload, their cardioprotective or cardiodestructive activities differ from each other. Among them, mast cells, neutrophils, and dendritic cells exert detrimental function in experimental models, whereas eosinophils and natural killer T cells display cardioprotective activities. Depending on their subsets, macrophages and monocytes may exacerbate cardiodysfunction or negatively regulate cardiac hypertrophy and remodeling. Pressure overload stimulates the secretion of cytokines, chemokines, and growth factors from innate immune cells and even resident cardiomyocytes that together assist innate immune cell infiltration into injured heart. These infiltrates are involved in pro-hypertrophic events and cardiac fibroblast activation. Immune regulation of cardiac innate immune cells becomes a promising therapeutic approach in experimental cardiac disease treatment, highlighting the significance of their clinical evaluation in humans.
Collapse
Affiliation(s)
- Xin Liu
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Guo-Ping Shi
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Junli Guo
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
- Hainan Provincial Key Laboratory for Tropical Cardiovascular Diseases Research & Key Laboratory of Emergency and Trauma of Ministry of Education, Institute of Cardiovascular Research of the First Affiliated Hospital, Hainan Medical University, Haikou, China
| |
Collapse
|
50
|
Howell KL, Kaji DA, Li TM, Montero A, Yeoh K, Nasser P, Huang AH. Macrophage depletion impairs neonatal tendon regeneration. FASEB J 2021; 35:e21618. [PMID: 33982337 DOI: 10.1096/fj.202100049r] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 03/26/2021] [Accepted: 04/09/2021] [Indexed: 12/21/2022]
Abstract
Tendons are dense connective tissues that transmit muscle forces to the skeleton. After adult injury, healing potential is generally poor and dominated by scar formation. Although the immune response is a key feature of healing, the specific immune cells and signals that drive tendon healing have not been fully defined. In particular, the immune regulators underlying tendon regeneration are almost completely unknown due to a paucity of tendon regeneration models. Using a mouse model of neonatal tendon regeneration, we screened for immune-related markers and identified upregulation of several genes associated with inflammation, macrophage chemotaxis, and TGFβ signaling after injury. Depletion of macrophages using AP20187 treatment of MaFIA mice resulted in impaired functional healing, reduced cell proliferation, reduced ScxGFP+ neo-tendon formation, and altered tendon gene expression. Collectively, these results show that inflammation is a key component of neonatal tendon regeneration and demonstrate a requirement for macrophages in effective functional healing.
Collapse
Affiliation(s)
- Kristen L Howell
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Deepak A Kaji
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Thomas M Li
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Angela Montero
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kenji Yeoh
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Philip Nasser
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alice H Huang
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|