1
|
Choudhary V, Choudhary M, Bollag WB. Exploring Skin Wound Healing Models and the Impact of Natural Lipids on the Healing Process. Int J Mol Sci 2024; 25:3790. [PMID: 38612601 PMCID: PMC11011291 DOI: 10.3390/ijms25073790] [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: 02/08/2024] [Revised: 03/21/2024] [Accepted: 03/23/2024] [Indexed: 04/14/2024] Open
Abstract
Cutaneous wound healing is a complex biological process involving a series of well-coordinated events aimed at restoring skin integrity and function. Various experimental models have been developed to study the mechanisms underlying skin wound repair and to evaluate potential therapeutic interventions. This review explores the diverse array of skin wound healing models utilized in research, ranging from rodent excisional wounds to advanced tissue engineering constructs and microfluidic platforms. More importantly, the influence of lipids on the wound healing process is examined, emphasizing their role in enhancing barrier function restoration, modulating inflammation, promoting cell proliferation, and promoting remodeling. Lipids, such as phospholipids, sphingolipids, and ceramides, play crucial roles in membrane structure, cell signaling, and tissue repair. Understanding the interplay between lipids and the wound microenvironment provides valuable insights into the development of novel therapeutic strategies for promoting efficient wound healing and tissue regeneration. This review highlights the significance of investigating skin wound healing models and elucidating the intricate involvement of lipids in the healing process, offering potential avenues for improving clinical outcomes in wound management.
Collapse
Affiliation(s)
- Vivek Choudhary
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (V.C.)
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA
| | - Mrunal Choudhary
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (V.C.)
| | - Wendy B. Bollag
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (V.C.)
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA
- Department of Dermatology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
2
|
Luo H, Wang Y, Su Y, Liu D, Xiao H, Wu M, Zhao Y, Xue F. Paracrine effects of adipose-derived stem cells in cutaneous wound healing in streptozotocin-induced diabetic rats. J Wound Care 2022; 31:S29-S38. [PMID: 35199561 DOI: 10.12968/jowc.2022.31.sup3.s29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
OBJECTIVE The purpose of this study was to explore the paracrine effects of adipose-derived stem cells (ASCs) on cutaneous wound healing in diabetic rats. METHOD The ASCs were isolated and identified by immunofluorescent staining. The ASCs-conditioned medium (ASCs-CM) was harvested. Cell counting kit (CCK)-8 assay, scratch experiments, western blot and quantitative polymerase chain reaction (qPCR) were performed to observe the effects of ASCs-CM on fibroblasts. A full-thickness skin wound diabetic rat model was prepared, using 34 male, Sprague Dawley rats. ASCs-CM or negative-control medium (N-CM) was injected around the wound surface. The existing wound area was measured on days 4, 8, 12 and 16 after the postoperative day, and the wound tissues were collected for immunohistochemical staining and qPCR quantitative study. RESULTS In this experiment, the isolated cells were characterised as ASCs. The results of CCK-8 assay, cell scratch test, western blot and qPCR showed ASCs-CM could significantly promote the proliferation, migration and differentiation of fibroblasts. Simultaneously, the healing rate of full-thickness skin wounds in diabetic rats was significantly higher in the ASCs-CM group than the N-CM group on days 4, 8, 12 and 16. Immunohistochemical staining and qPCR results showed that the expression of vascular endothelial growth factor (VEGF, days 4 and 8), α-smooth muscle actin (SMA) (days 4 and 16), transforming growth factor (TGF)-β1 (days 4, 8 and 12) were higher in the ASCs-CM group than that of the N-CM group (p<0.05). CONCLUSION This experiment demonstrated that ASCs-CM may accelerate wound healing in diabetic rats by promoting the secretion of TGF-β1, VEGF and the proliferation, migration and differentiation of fibroblasts.
Collapse
Affiliation(s)
- Hua Luo
- Department of Orthopaedics, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou 317000, Zhejiang, China.,Fengxian District Central Hospital Graduate Student Training Base, Jinzhou Medical University, Shanghai 201499, China.,Department of Laboratory Medicine & Central Laboratory, Fengxian District Central Hospital, Shanghai 201499, China
| | - Yongjian Wang
- Department of Orthopaedics, The First People's Hospital of Wenling, Taizhou, Zhejiang 317500, China
| | - Yongwei Su
- The First Affiliated Hospital of Jinzhou Medical University, Graduate Student Training Base, Jinzhou Medical University, Jinzhou 121000, Liaoning, China
| | - Danping Liu
- The First Affiliated Hospital of Jinzhou Medical University, Graduate Student Training Base, Jinzhou Medical University, Jinzhou 121000, Liaoning, China
| | - Haijun Xiao
- Department of Orthopaedics, Fengxian District Central Hospital, Shanghai 201499, China
| | - Ming Wu
- Department of Orthopaedics, Fengxian District Central Hospital, Shanghai 201499, China
| | - Yong Zhao
- Department of Orthopaedics, Fengxian District Central Hospital, Shanghai 201499, China
| | - Feng Xue
- Fengxian District Central Hospital Graduate Student Training Base, Jinzhou Medical University, Shanghai 201499, China.,Department of Orthopaedics, Fengxian District Central Hospital, Shanghai 201499, China
| |
Collapse
|
3
|
Ning M, Li Y, Chen Z, Han P, Tang X, Zhang Y, Gao L. Role of haematopoietic cell-specific protein 1-associated protein X-1 gene in lipopolysaccharide-induced apoptosis of human dermal fibroblasts. Wound Repair Regen 2021; 30:34-44. [PMID: 34826355 DOI: 10.1111/wrr.12985] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 09/18/2021] [Accepted: 11/01/2021] [Indexed: 12/11/2022]
Abstract
Wound healing may be disrupted by lipopolysaccharide (LPS)-induced mitochondrial dysfunction, inflammation, and excessive oxidative stress, which can lead to undesirable consequences. The haematopoietic cell-specific protein 1-associated protein X-1 (HAX-1) is a mitochondrial matrix protein that regulates mitochondrial function. This study aimed to comprehensively identify the role of HAX-1 in the inhibition of LPS-induced mitochondrial dysfunction and apoptosis in human dermal fibroblasts (HDFs). HAX-1 expression was assessed in the HDF-a cell line using real-time polymerase chain reaction, western blotting, and immunohistochemical staining. The viability, migration, and apoptosis of HDF-a cells were evaluated using the water-soluble tetrazolium-1 assay, transwell assay, and flow cytometry analysis, respectively. Mitochondrial function was evaluated based on reactive oxygen species (ROS) generation and mitochondrial membrane potential (ΔΨm). Our results demonstrated that LPS stimulation markedly repressed HAX-1 expression in HDFs and silencing of HAX-1 led to mitochondrial ROS accumulation, ΔΨm disruption, and abnormal mitochondrial morphology. Accordingly, overexpression of HAX-1 or administration of metformin enhanced mitochondrial fusion and normalized mitochondrial dynamics, thereby reversing LPS-induced mitochondrial dysfunction, fibroblast apoptosis, and viability and migration inhibition in HDF-a cells. These data support a mechanism wherein HAX-1 plays a crucial role in LPS-induced fibroblast apoptosis in a mitochondria-dependent manner.
Collapse
Affiliation(s)
- Mei Ning
- Department of General Surgery, Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang, China
| | - Yanlong Li
- Department of Cardiovascular, Shandong Electric Power Central Hospital, Jinan, China
| | - Zhihui Chen
- Department of Emergency, Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang, China
| | - Pengfei Han
- Department of Clinical Laboratory, Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang, China
| | - Xiaolan Tang
- College of Nursing, Mudanjiang Medical University, Mudanjiang, China
| | - Yanli Zhang
- Department of Anesthesiology, Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang, China
| | - Luan Gao
- Department of Emergency, Hongqi Hospital Affiliated to Mudanjiang Medical University, Mudanjiang, China
| |
Collapse
|
4
|
Pils V, Terlecki-Zaniewicz L, Schosserer M, Grillari J, Lämmermann I. The role of lipid-based signalling in wound healing and senescence. Mech Ageing Dev 2021; 198:111527. [PMID: 34174292 DOI: 10.1016/j.mad.2021.111527] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 05/28/2021] [Accepted: 06/17/2021] [Indexed: 02/07/2023]
Abstract
Lipid-based signalling modulates several cellular processes and intercellular communication during wound healing and tissue regeneration. Bioactive lipids include but are not limited to the diverse group of eicosanoids, phospholipids, and extracellular vesicles and mediate the attraction of immune cells, initiation of inflammatory responses, and their resolution. In aged individuals, wound healing and tissue regeneration are greatly impaired, resulting in a delayed healing process and non-healing wounds. Senescent cells accumulate with age in vivo, preferably at sites implicated in age-associated pathologies and their elimination was shown to alleviate many age-associated diseases and disorders. In contrast to these findings, the transient presence of senescent cells in the process of wound healing exerts beneficial effects and limits fibrosis. Hence, clearance of senescent cells during wound healing was repeatedly shown to delay wound closure in vivo. Recent findings established a dysregulated synthesis of eicosanoids, phospholipids and extracellular vesicles as part of the senescent phenotype. This intriguing connection between cellular senescence, lipid-based signalling, and the process of wound healing and tissue regeneration prompts us to compile the current knowledge in this review and propose future directions for investigation.
Collapse
Affiliation(s)
- Vera Pils
- Christian Doppler Laboratory for the Biotechnology of Skin Aging, Institute of Molecular Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Lucia Terlecki-Zaniewicz
- Christian Doppler Laboratory for the Biotechnology of Skin Aging, Institute of Molecular Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Markus Schosserer
- Christian Doppler Laboratory for Skin Multimodal Imaging of Aging and Senescence - SKINMAGINE, Institute of Molecular Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria; Austrian Cluster for Tissue Regeneration, Austria
| | - Johannes Grillari
- Christian Doppler Laboratory for the Biotechnology of Skin Aging, Institute of Molecular Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria; Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Linz and Vienna, Austria; Austrian Cluster for Tissue Regeneration, Austria
| | - Ingo Lämmermann
- Christian Doppler Laboratory for the Biotechnology of Skin Aging, Institute of Molecular Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria.
| |
Collapse
|
5
|
Systems biology predicts that fibrosis in tuberculous granulomas may arise through macrophage-to-myofibroblast transformation. PLoS Comput Biol 2020; 16:e1008520. [PMID: 33370784 PMCID: PMC7793262 DOI: 10.1371/journal.pcbi.1008520] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 01/08/2021] [Accepted: 11/11/2020] [Indexed: 02/07/2023] Open
Abstract
Mycobacterium tuberculosis (Mtb) infection causes tuberculosis (TB), a disease characterized by development of granulomas. Granulomas consist of activated immune cells that cluster together to limit bacterial growth and restrict dissemination. Control of the TB epidemic has been limited by lengthy drug regimens, antibiotic resistance, and lack of a robustly efficacious vaccine. Fibrosis commonly occurs during treatment and is associated with both positive and negative disease outcomes in TB but little is known about the processes that initiate fibrosis in granulomas. Human and nonhuman primate granulomas undergoing fibrosis can have spindle-shaped macrophages with fibroblast-like morphologies suggesting a relationship between macrophages, fibroblasts, and granuloma fibrosis. This relationship has been difficult to investigate because of the limited availability of human pathology samples, the time scale involved in human TB, and overlap between fibroblast and myeloid cell markers in tissues. To better understand the origins of fibrosis in TB, we used a computational model of TB granuloma biology to identify factors that drive fibrosis over the course of local disease progression. We validated the model with granulomas from nonhuman primates to delineate myeloid cells and lung-resident fibroblasts. Our results suggest that peripheral granuloma fibrosis, which is commonly observed, can arise through macrophage-to-myofibroblast transformation (MMT). Further, we hypothesize that MMT is induced in M1 macrophages through a sequential combination of inflammatory and anti-inflammatory signaling in granuloma macrophages. We predict that MMT may be a mechanism underlying granuloma-associated fibrosis and warrants further investigation into myeloid cells as drivers of fibrotic disease. Tuberculosis is a disease caused by Mycobacterium tuberculosis (Mtb), a bacterium that infects over a third of the world’s population. The only available vaccine for TB has limited efficacy and drug treatment involves several antibiotics that are taken for several months. These drugs can have significant side-effects and a lack of compliance can lead to drug resistance in Mtb. A hallmark of Mtb infection is the development of clusters of cells that form around infected macrophages to contain the infection called granulomas. Older granulomas, or granulomas in patients treated with antibiotic, often become fibrotic and this can cause chronic lung problems long after the Mtb infection has cleared. The process that drives a fibrotic outcome has been difficult to assess in vivo. Herein we combined wet-lab and computational experimentation to identify a novel mechanism leading to peripheral fibrosis in granulomas. We find that macrophages transforming into myofibroblast-like cells, may be a key pathway to granuloma-associated fibrosis, a phenomena that has not been well characterized in vivo. Further we identified factors that can inhibit fibrosis development during TB that could be therapeutic targets during TB treatment to limit the risk of long-term tissue damage in TB.
Collapse
|
6
|
Leonard-Duke J, Evans S, Hannan RT, Barker TH, Bates JHT, Bonham CA, Moore BB, Kirschner DE, Peirce SM. Multi-scale models of lung fibrosis. Matrix Biol 2020; 91-92:35-50. [PMID: 32438056 DOI: 10.1016/j.matbio.2020.04.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 03/13/2020] [Accepted: 04/15/2020] [Indexed: 02/08/2023]
Abstract
The architectural complexity of the lung is crucial to its ability to function as an organ of gas exchange; the branching tree structure of the airways transforms the tracheal cross-section of only a few square centimeters to a blood-gas barrier with a surface area of tens of square meters and a thickness on the order of a micron or less. Connective tissue comprised largely of collagen and elastic fibers provides structural integrity for this intricate and delicate system. Homeostatic maintenance of this connective tissue, via a balance between catabolic and anabolic enzyme-driven processes, is crucial to life. Accordingly, when homeostasis is disrupted by the excessive production of connective tissue, lung function deteriorates rapidly with grave consequences leading to chronic lung conditions such as pulmonary fibrosis. Understanding how pulmonary fibrosis develops and alters the link between lung structure and function is crucial for diagnosis, prognosis, and therapy. Further information gained could help elaborate how the healing process breaks down leading to chronic disease. Our understanding of fibrotic disease is greatly aided by the intersection of wet lab studies and mathematical and computational modeling. In the present review we will discuss how multi-scale modeling has facilitated our understanding of pulmonary fibrotic disease as well as identified opportunities that remain open and have produced techniques that can be incorporated into this field by borrowing approaches from multi-scale models of fibrosis beyond the lung.
Collapse
Affiliation(s)
- Julie Leonard-Duke
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Stephanie Evans
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Riley T Hannan
- Department of Pathology, University of Virginia, Charlottesville, VA 22908, USA
| | - Thomas H Barker
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Jason H T Bates
- Department of Medicine, Vermont Lung Center, University of Vermont College of Medicine, Burlington, VT 05405, USA
| | - Catherine A Bonham
- Division of Pulmonary and Critical Care Medicine, University of Virginia, Charlottesville VA 22908, USA
| | - Bethany B Moore
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, and Department of Microbiology and Immunology, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA
| | - Denise E Kirschner
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Shayn M Peirce
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA; Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
7
|
Li Z, Zhang J, Zhou J, Lu L, Wang H, Zhang G, Wan G, Cai S, Du J. Nodal Facilitates Differentiation of Fibroblasts to Cancer-Associated Fibroblasts that Support Tumor Growth in Melanoma and Colorectal Cancer. Cells 2019; 8:E538. [PMID: 31167491 PMCID: PMC6627322 DOI: 10.3390/cells8060538] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 06/02/2019] [Accepted: 06/03/2019] [Indexed: 02/07/2023] Open
Abstract
Fibroblasts become cancer-associated fibroblasts (CAFs) in the tumor microenvironment after activation by transforming growth factor-β (TGF-β) and are critically involved in cancer progression. However, it is unknown whether the TGF superfamily member Nodal, which is expressed in various tumors but not expressed in normal adult tissue, influences the fibroblast to CAF conversion. Here, we report that Nodal has a positive correlation with α-smooth muscle actin (α-SMA) in clinical melanoma and colorectal cancer (CRC) tissues. We show the Nodal converts normal fibroblasts to CAFs, together with Snail and TGF-β signaling pathway activation in fibroblasts. Activated CAFs promote cancer growth in vitro and tumor-bearing mouse models in vivo. These results demonstrate that intercellular crosstalk between cancer cells and fibroblasts is mediated by Nodal, which controls tumor growth, providing potential targets for the prevention and treatment of tumors.
Collapse
Affiliation(s)
- Ziqian Li
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| | - Junjie Zhang
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| | - Jiawang Zhou
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| | - Linlin Lu
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| | - Hongsheng Wang
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| | - Ge Zhang
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| | - Guohui Wan
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| | - Shaohui Cai
- Department of Pharmacology, School of Pharmaceutical Sciences, Jinan University, Guangzhou 510632, China.
| | - Jun Du
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| |
Collapse
|
8
|
Zhou ZQ, Chen Y, Chai M, Tao R, Lei YH, Jia YQ, Shu J, Ren J, Li G, Wei WX, Han YD, Han Y. Adipose extracellular matrix promotes skin wound healing by inducing the differentiation of adipose‑derived stem cells into fibroblasts. Int J Mol Med 2019; 43:890-900. [PMID: 30535488 PMCID: PMC6317660 DOI: 10.3892/ijmm.2018.4006] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 11/20/2018] [Indexed: 12/25/2022] Open
Abstract
Fibroblasts are the major effector cells of skin wound healing. Adipose‑derived stem cells can differentiate into fibroblasts under certain conditions. In the present study, it was hypothesized that adipose‑derived stem cells (ADSCs) could be induced by the adipose extracellular matrix (ECM) to differentiate into fibroblasts in order to promote skin wound healing. First, flow cytometry was used to detect the ratio of fibroblasts and relative expression of the fibroblast markers cytokeratin 19 (CK19) and vimentin in ADSCs. Then, the effect of the adipose ECM during the differentiation of ADSCs into fibroblasts was investigated by detecting the total amount of collagen fibers and degree of fibrosis, and the proliferation and cell cycle of differentiated fibroblasts, using the MTT assay and flow cytometry analysis respectively. Finally, a mouse skin wound model was established and treated with PBS, ADSC suspension or ECM + ADSCs to compare wound healing rate and expression of collagen I and collagen III by immunohistochemistry. Following induction of ADSCs with the adipose ECM, more fibroblasts were found, expression of CK19 and vimentin increased, and a greater degree of fibrosis occurred, which revealed the positive effect of the adipose ECM on the differentiation of ADSCs into fibroblasts. In addition, the induced fibroblasts had enhanced proliferation activity, with more cells in the S phase and fewer in the G2/M phase. The in vivo experiment indicated that the ECM produced by the ADSCs had a faster wound healing rate and increased expression of collagen I and collagen III compared with mice injected with PBS or ADSCs alone, which verified that ADSCs induced by the adipose ECM had a positive effect on skin wound healing. The present study demonstrated that the adipose ECM in combination with ADSCs may be a novel therapeutic target for the repair of skin injury, due to the ability of the adipose ECM to induce the differentiation of ADSCs into fibroblasts and to facilitate the wound healing process.
Collapse
Affiliation(s)
- Zhi-Qiang Zhou
- Department of Plastic and Reconstructive Surgery, General Hospital of Chinese PLA, Beijing 100853
| | - Yi Chen
- Institute of Bioengineering, Academy of Military Medical Research, Academy of Military Science of Chinese PLA, Beijing 100071, P.R. China
| | - Mi Chai
- Department of Plastic and Reconstructive Surgery, General Hospital of Chinese PLA, Beijing 100853
| | - Ran Tao
- Department of Plastic and Reconstructive Surgery, General Hospital of Chinese PLA, Beijing 100853
| | - Yong-Hong Lei
- Department of Plastic and Reconstructive Surgery, General Hospital of Chinese PLA, Beijing 100853
| | - Yi-Qing Jia
- Department of Plastic and Reconstructive Surgery, General Hospital of Chinese PLA, Beijing 100853
| | - Jun Shu
- Department of Plastic and Reconstructive Surgery, General Hospital of Chinese PLA, Beijing 100853
| | - Jing Ren
- Department of Plastic and Reconstructive Surgery, General Hospital of Chinese PLA, Beijing 100853
| | - Guo Li
- Department of Plastic and Reconstructive Surgery, General Hospital of Chinese PLA, Beijing 100853
| | - Wen-Xin Wei
- Department of Plastic and Reconstructive Surgery, General Hospital of Chinese PLA, Beijing 100853
| | - Yu-Di Han
- Department of Plastic and Reconstructive Surgery, General Hospital of Chinese PLA, Beijing 100853
| | - Yan Han
- Department of Plastic and Reconstructive Surgery, General Hospital of Chinese PLA, Beijing 100853
| |
Collapse
|
9
|
Chen MJ, Whiteley JP, Please CP, Ehlicke F, Waters SL, Byrne HM. Identifying chondrogenesis strategies for tissue engineering of articular cartilage. J Tissue Eng 2019; 10:2041731419842431. [PMID: 31040937 PMCID: PMC6481001 DOI: 10.1177/2041731419842431] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 03/14/2019] [Indexed: 11/17/2022] Open
Abstract
A key step in the tissue engineering of articular cartilage is the chondrogenic differentiation of mesenchymal stem cells (MSCs) into chondrocytes (native cartilage cells). Chondrogenesis is regulated by transforming growth factor-β (TGF-β), a short-lived cytokine whose effect is prolonged by storage in the extracellular matrix. Tissue engineering applications aim to maximise the yield of differentiated MSCs. Recent experiments involve seeding a hydrogel construct with a layer of MSCs lying below a layer of chondrocytes, stimulating the seeded cells in the construct from above with exogenous TGF-β and then culturing it in vitro. To investigate the efficacy of this strategy, we develop a mathematical model to describe the interactions between MSCs, chondrocytes and TGF-β. Using this model, we investigate the effect of varying the initial concentration of TGF-β, the initial densities of the MSCs and chondrocytes, and the relative depths of the two layers on the long-time composition of the tissue construct.
Collapse
Affiliation(s)
- Michael J Chen
- School of Mathematical Sciences, The University of Adelaide, Adelaide, SA, Australia
- Mathematical Institute, University of Oxford, Oxford, UK
| | | | - Colin P Please
- Mathematical Institute, University of Oxford, Oxford, UK
| | - Franziska Ehlicke
- Department of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Sarah L Waters
- Mathematical Institute, University of Oxford, Oxford, UK
| | - Helen M Byrne
- Mathematical Institute, University of Oxford, Oxford, UK
| |
Collapse
|
10
|
Cicchese JM, Evans S, Hult C, Joslyn LR, Wessler T, Millar JA, Marino S, Cilfone NA, Mattila JT, Linderman JJ, Kirschner DE. Dynamic balance of pro- and anti-inflammatory signals controls disease and limits pathology. Immunol Rev 2018; 285:147-167. [PMID: 30129209 PMCID: PMC6292442 DOI: 10.1111/imr.12671] [Citation(s) in RCA: 204] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Immune responses to pathogens are complex and not well understood in many diseases, and this is especially true for infections by persistent pathogens. One mechanism that allows for long-term control of infection while also preventing an over-zealous inflammatory response from causing extensive tissue damage is for the immune system to balance pro- and anti-inflammatory cells and signals. This balance is dynamic and the immune system responds to cues from both host and pathogen, maintaining a steady state across multiple scales through continuous feedback. Identifying the signals, cells, cytokines, and other immune response factors that mediate this balance over time has been difficult using traditional research strategies. Computational modeling studies based on data from traditional systems can identify how this balance contributes to immunity. Here we provide evidence from both experimental and mathematical/computational studies to support the concept of a dynamic balance operating during persistent and other infection scenarios. We focus mainly on tuberculosis, currently the leading cause of death due to infectious disease in the world, and also provide evidence for other infections. A better understanding of the dynamically balanced immune response can help shape treatment strategies that utilize both drugs and host-directed therapies.
Collapse
Affiliation(s)
- Joseph M. Cicchese
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Stephanie Evans
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Caitlin Hult
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Louis R. Joslyn
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Timothy Wessler
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Jess A. Millar
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Simeone Marino
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Nicholas A. Cilfone
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Joshua T. Mattila
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Denise E. Kirschner
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
11
|
Chen MJ, Whiteley JP, Please CP, Schwab A, Ehlicke F, Waters SL, Byrne HM. Inducing chondrogenesis in MSC/chondrocyte co-cultures using exogenous TGF-β: a mathematical model. J Theor Biol 2018; 439:1-13. [DOI: 10.1016/j.jtbi.2017.11.024] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 09/22/2017] [Accepted: 11/30/2017] [Indexed: 11/30/2022]
|
12
|
Warsinske HC, Pienaar E, Linderman JJ, Mattila JT, Kirschner DE. Deletion of TGF-β1 Increases Bacterial Clearance by Cytotoxic T Cells in a Tuberculosis Granuloma Model. Front Immunol 2017; 8:1843. [PMID: 29326718 PMCID: PMC5742530 DOI: 10.3389/fimmu.2017.01843] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 12/06/2017] [Indexed: 01/10/2023] Open
Abstract
Mycobacterium tuberculosis is the pathogenic bacterium that causes tuberculosis (TB), one of the most lethal infectious diseases in the world. The only vaccine against TB is minimally protective, and multi-drug resistant TB necessitates new therapeutics to treat infection. Developing new therapies requires a better understanding of the complex host immune response to infection, including dissecting the processes leading to formation of granulomas, the dense cellular lesions associated with TB. In this work, we pair experimental and computational modeling studies to explore cytokine regulation in the context of TB. We use our next-generation hybrid multi-scale model of granuloma formation (GranSim) to capture molecular, cellular, and tissue scale dynamics of granuloma formation. We identify TGF-β1 as a major inhibitor of cytotoxic T-cell effector function in granulomas. Deletion of TGF-β1 from the system results in improved bacterial clearance and lesion sterilization. We also identify a novel dichotomous regulation of cytotoxic T cells and macrophages by TGF-β1 and IL-10, respectively. These findings suggest that increasing cytotoxic T-cell effector functions may increase bacterial clearance in granulomas and highlight potential new therapeutic targets for treating TB.
Collapse
Affiliation(s)
- Hayley C Warsinske
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Elsje Pienaar
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, United States.,Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Jennifer J Linderman
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Joshua T Mattila
- Department of Infectious Diseases and Microbiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, United States
| | - Denise E Kirschner
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
13
|
Dong J, Yuan X, Xie W. Pentoxifylline exerts anti-inflammatory effects on cerebral ischemia reperfusion‑induced injury in a rat model via the p38 mitogen-activated protein kinase signaling pathway. Mol Med Rep 2017; 17:1141-1147. [PMID: 29115594 DOI: 10.3892/mmr.2017.7953] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 07/27/2017] [Indexed: 11/06/2022] Open
Abstract
Pentoxifylline exhibits complex functions with extensive pharmacological effects and is used therapeutically due to its therapeutic effects and rapid metabolism in the body, with no cumulative effects and few side effects. The present study investigated the effects of pentoxifylline on cerebral ischemia reperfusion‑induced injury (IRI) through suppression of inflammation in rats. Hematoxylin and eosin staining was performed to evaluate the number of neurocytes, and ELISAs were applied to measure tumor necrosis factor‑α, interleukin‑6, malondialdehyde and superoxide dismutase activities. Treatment with pentoxifylline significantly recovered the cerebral ischemia reperfusion‑induced neurological deficit score and cerebral infarct volume in rats. In addition, pentoxifylline treatment significantly reversed the cerebral ischemia reperfusion‑induced interleukin‑6, tumor necrosis factor‑α, malondialdehyde and superoxide dismutase levels in vivo. Furthermore, pentoxifylline significantly inhibited cyclooxygenase‑2 and inducible nitric oxide synthase mRNA and protein expression in cerebral IRI mice. Treatment with pentoxifylline also significantly suppressed the expression of cleaved caspase‑3 and p38 mitogen‑activated protein kinase (MAPK) protein in cerebral IRI mice. These results indicate that the protective effects of pentoxifylline on cerebral IRI may occur via the p38 MAPK signaling pathway.
Collapse
Affiliation(s)
- Jianqing Dong
- Department of Blood Transfusion, Zhangqiu People's Hospital, Jinan, Shandong 250200, P.R. China
| | - Xinmei Yuan
- Department of Blood Transfusion, Zhangqiu People's Hospital, Jinan, Shandong 250200, P.R. China
| | - Weiwei Xie
- Department of Blood Transfusion, Zhangqiu People's Hospital, Jinan, Shandong 250200, P.R. China
| |
Collapse
|
14
|
Zhu Z, Yi S, Shan Z, Guo H, Ke S. Effect of isoflurane + N 2O inhalation and propofol + fentanyl anesthesia on myocardial function as assessed by cardiac troponin, caspase-3, cyclooxygenase-2 and inducible nitric oxide synthase expression. Exp Ther Med 2017; 14:4377-4382. [PMID: 29067116 DOI: 10.3892/etm.2017.5029] [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: 02/21/2016] [Accepted: 05/11/2017] [Indexed: 11/06/2022] Open
Abstract
The aim of this study was to evaluate the effect of isoflurane + N2O inhalation and propofol + fentanyl anesthesia on myocardial function as assessed by cardiac troponin T (cTnT). A total of 60 patients were randomized into two groups: isoflurane + N2O inhalation (n=30) and propofol + fentanyl anesthesia (n=30). The findings demonstrated that there was no significant difference between the two experimental groups in terms of cTnT levels, demographic properties or hemodynamic parameters. Isoflurane + N2O inhalation and propofol + fentanyl anesthesia, respectively, were also investigated in a rat model of myocardial infarction. Myocardial cell damage, inflammation and oxidative stress levels, caspase-3/9 activities and cyclooxygenase-2 protein expression were markedly decreased, although there was no statistical significance difference between the two experimental groups. Notably, inducible nitric oxide synthase protein expression in the isoflurane + N2O inhalation group was significantly higher than that of the propofol + fentanyl anesthesia group (P<0.01). In conclusion, isoflurane + N2O inhalation and propofol + fentanyl anesthesia are not associated with risks for myocardial function.
Collapse
Affiliation(s)
- Zhuanghui Zhu
- Department of Cardiac Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, P.R. China
| | - Shuanglian Yi
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, Xiamen, Fujian 361003, P.R. China
| | - Zhonggui Shan
- Department of Cardiac Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, P.R. China.,Department of Cardiac Surgery, Teaching Hospital of Fujian Medical University, Xiamen, Fujian 361003, P.R. China
| | - Hongwei Guo
- Department of Cardiac Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, P.R. China
| | - Shaofan Ke
- Department of Cardiac Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, P.R. China
| |
Collapse
|
15
|
Warsinske HC, DiFazio RM, Linderman JJ, Flynn JL, Kirschner DE. Identifying mechanisms driving formation of granuloma-associated fibrosis during Mycobacterium tuberculosis infection. J Theor Biol 2017. [PMID: 28642013 DOI: 10.1016/j.jtbi.2017.06.017] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB), is a pulmonary pathogen of major global concern. A key feature of Mtb infection in primates is the formation of granulomas, dense cellular structures surrounding infected lung tissue. These structures serve as the main site of host-pathogen interaction in TB, and thus to effectively treat TB we must clarify mechanisms of granuloma formation and their function in disease. Fibrotic granulomas are associated with both good and bad disease outcomes. Fibrosis can serve to isolate infected tissue from healthy tissue, but it can also cause difficulty breathing as it leaves scars. Little is known about fibrosis in TB, and data from non-human primates is just beginning to clarify the picture. This work focuses on constructing a hybrid multi-scale model of fibrotic granuloma formation, in order to identify mechanisms driving development of fibrosis in Mtb infected lungs. We combine dynamics of molecular, cellular, and tissue scale models from previously published studies to characterize the formation of two common sub-types of fibrotic granulomas: peripherally fibrotic, with a cuff of collagen surrounding granulomas, and centrally fibrotic, with collagen throughout granulomas. Uncertainty and sensitivity analysis, along with large simulation sets, enable us to identify mechanisms differentiating centrally versus peripherally fibrotic granulomas. These findings suggest that heterogeneous cytokine environments exist within granulomas and may be responsible for driving tissue scale morphologies. Using this model we are primed to better understand the complex structure of granulomas, a necessity for developing successful treatments for TB.
Collapse
Affiliation(s)
- Hayley C Warsinske
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, United States of America
| | - Robert M DiFazio
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, United States of America
| | - Jennifer J Linderman
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, United States of America
| | - JoAnne L Flynn
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261, United States of America
| | - Denise E Kirschner
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, United States of America.
| |
Collapse
|
16
|
Warsinske HC, Wheaton AK, Kim KK, Linderman JJ, Moore BB, Kirschner DE. Computational Modeling Predicts Simultaneous Targeting of Fibroblasts and Epithelial Cells Is Necessary for Treatment of Pulmonary Fibrosis. Front Pharmacol 2016; 7:183. [PMID: 27445819 PMCID: PMC4917547 DOI: 10.3389/fphar.2016.00183] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 06/10/2016] [Indexed: 11/13/2022] Open
Abstract
Pulmonary fibrosis is pathologic remodeling of lung tissue that can result in difficulty breathing, reduced quality of life, and a poor prognosis for patients. Fibrosis occurs as a result of insult to lung tissue, though mechanisms of this response are not well-characterized. The disease is driven in part by dysregulation of fibroblast proliferation and differentiation into myofibroblast cells, as well as pro-fibrotic mediator-driven epithelial cell apoptosis. The most well-characterized pro-fibrotic mediator associated with pulmonary fibrosis is TGF-β1. Excessive synthesis of, and sensitivity to, pro-fibrotic mediators as well as insufficient production of and sensitivity to anti-fibrotic mediators has been credited with enabling fibroblast accumulation. Available treatments neither halt nor reverse lung damage. In this study we have two aims: to identify molecular and cellular scale mechanisms driving fibroblast proliferation and differentiation as well as epithelial cell survival in the context of fibrosis, and to predict therapeutic targets and strategies. We combine in vitro studies with a multi-scale hybrid agent-based computational model that describes fibroblasts and epithelial cells in co-culture. Within this model TGF-β1 represents a pro-fibrotic mediator and we include detailed dynamics of TGF-β1 receptor ligand signaling in fibroblasts. PGE2 represents an anti-fibrotic mediator. Using uncertainty and sensitivity analysis we identify TGF-β1 synthesis, TGF-β1 activation, and PGE2 synthesis among the key mechanisms contributing to fibrotic outcomes. We further demonstrate that intervention strategies combining potential therapeutics targeting both fibroblast regulation and epithelial cell survival can promote healthy tissue repair better than individual strategies. Combinations of existing drugs and compounds may provide significant improvements to the current standard of care for pulmonary fibrosis. Thus, a two-hit therapeutic intervention strategy may prove necessary to halt and reverse disease dynamics.
Collapse
Affiliation(s)
- Hayley C. Warsinske
- Department of Microbiology and Immunology, University of Michigan Medical SchoolAnn Arbor, MI, USA
| | - Amanda K. Wheaton
- Department of Internal Medicine, University of Michigan Medical SchoolAnn Arbor, MI, USA
| | - Kevin K. Kim
- Department of Internal Medicine, University of Michigan Medical SchoolAnn Arbor, MI, USA
| | | | - Bethany B. Moore
- Department of Microbiology and Immunology, University of Michigan Medical SchoolAnn Arbor, MI, USA
- Department of Internal Medicine, University of Michigan Medical SchoolAnn Arbor, MI, USA
| | - Denise E. Kirschner
- Department of Microbiology and Immunology, University of Michigan Medical SchoolAnn Arbor, MI, USA
| |
Collapse
|