Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
J Invest Dermatol ; 2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38580106

ABSTRACT

Systemic sclerosis (SSc) is a challenging autoimmune disease characterized by progressive fibrosis affecting the skin and internal organs. Despite the known infiltration of macrophages and neutrophils, their precise contributions to SSc pathogenesis remain elusive. In this study, we elucidated that CD206hiMHCIIlo M2-like macrophages constitute the predominant pathogenic immune cell population in the fibrotic skin of a bleomycin-induced SSc mouse model. These cells emerged as pivotal contributors to the profibrotic response by orchestrating the production of TGF-ß1 through a MerTK signaling-dependent manner. Notably, we observed that neutrophil infiltration was a prerequisite for accumulation of M2-like macrophages. Strategies such as neutrophil depletion or inhibition of CXCR1/2 were proven effective in reducing M2-like macrophages, subsequently mitigating SSc progression. Detailed investigations revealed that in fibrotic skin, neutrophil-released neutrophil extracellular traps were responsible for the differentiation of M2-like macrophages. Our findings illuminate the significant involvement of the neutrophil-macrophage-fibrosis axis in SSc pathogenesis, offering critical information for the development of potential therapeutic strategies.

2.
Mol Ther ; 32(4): 1144-1157, 2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38310354

ABSTRACT

The potent immunomodulatory function of mesenchymal stem/stromal cells (MSCs) elicited by proinflammatory cytokines IFN-γ and TNF-α (IT) is critical to resolve inflammation and promote tissue repair. However, little is known about how the immunomodulatory capability of MSCs is related to their differentiation competency in the inflammatory microenvironment. In this study, we demonstrate that the adipocyte differentiation and immunomodulatory function of human adipose tissue-derived MSCs (MSC(AD)s) are mutually exclusive. Mitochondrial reactive oxygen species (mtROS), which promote adipocyte differentiation, were decreased in MSC(AD)s due to IT-induced upregulation of superoxide dismutase 2 (SOD2). Furthermore, knockdown of SOD2 led to enhanced adipogenic differentiation but reduced immunosuppression capability of MSC(AD)s. Interestingly, the adipogenic differentiation was associated with increased mitochondrial biogenesis and upregulation of peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PPARGC1A/PGC-1α) expression. IT inhibited PGC-1α expression and decreased mitochondrial mass but promoted glycolysis in an SOD2-dependent manner. MSC(AD)s lacking SOD2 were compromised in their therapeutic efficacy in DSS-induced colitis in mice. Taken together, these findings indicate that the adipogenic differentiation and immunomodulation of MSC(AD)s may compete for resources in fulfilling the respective biosynthetic needs. Blocking of adipogenic differentiation by mitochondrial antioxidant may represent a novel strategy to enhance the immunosuppressive activity of MSCs in the inflammatory microenvironment.


Subject(s)
Mesenchymal Stem Cells , Superoxide Dismutase , Mice , Humans , Animals , Cell Differentiation , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Adipocytes , Mesenchymal Stem Cells/metabolism
3.
Cell Death Dis ; 15(1): 70, 2024 01 18.
Article in English | MEDLINE | ID: mdl-38238297

ABSTRACT

Glucocorticoid (GC) is essential for maintaining immune homeostasis. While GC is known to regulate the expression of genes related to inflammation in immune cells, the effects of GC, especially in the presence of inflammation, on non-immune cells remain largely unexplored. In particular, the impact of GC on inflammatory cytokine-induced immune modulatory responses of tissue stromal cells is unknown, though it has been widely used to modulate tissue injuries. Here we found that GC could enhance the expression of TSG6, a vital tissue repair effector molecule, in IFNγ and TNFα treated human umbilical cord (UC)-MSCs. NF-κB activation was found to be required for GC-augmented TSG6 upregulation. STAT3, but not STAT1, was also found to be required for the TSG6 upregulation in MSCs exposed to IFNγ, TNFα and GC. Moreover, the phosphorylation (activation) of STAT3 was attenuated when NF-κB was knocked down. Importantly, human UC-MSCs pretreated with a cocktail containing GC, IFNγ, and TNFα could significantly enhance the therapeutic effect of human UC-MSCs in an acute lung injury mouse model, as reflected by reduced infiltration of immune cells and down-regulation of iNOS in macrophages in the lung. Together, the findings reveal a novel link between GR, NF-κB and STAT3 in regulating the immunomodulatory and regenerative properties of MSCs, providing novel information for the understanding and treatment of inflammatory conditions.


Subject(s)
Mesenchymal Stem Cells , NF-kappa B , Mice , Animals , Humans , NF-kappa B/metabolism , Cytokines/metabolism , Glucocorticoids/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/metabolism , Inflammation/metabolism , Mesenchymal Stem Cells/metabolism , STAT3 Transcription Factor/metabolism
4.
Mol Oncol ; 2023 Nov 12.
Article in English | MEDLINE | ID: mdl-37953485

ABSTRACT

Hyaluronic acid (HA) is a major component of the extracellular matrix, providing essential mechanical scaffolding for cells and, at the same time, mediating essential biochemical signals required for tissue homeostasis. Many solid tumors are characterized by dysregulated HA metabolism, resulting in increased HA levels in cancer tissues. HA interacts with several cell surface receptors, such as cluster of differentiation 44 and receptor for hyaluronan-mediated motility, thus co-regulating important signaling pathways in cancer development and progression. In this review, we describe the enzymes controlling HA metabolism and its intracellular effectors emphasizing their impact on cancer chemotherapy resistance. We will also explore the current and future prospects of HA-based therapy, highlighting the opportunities and challenges in the field.

5.
Oncogene ; 42(46): 3371-3384, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37848625

ABSTRACT

Epithelial tissue homeostasis is closely associated with the self-renewal and differentiation behaviors of epithelial stem cells (ESCs). p63, a well-known marker of ESCs, is an indispensable factor for their biological activities during epithelial development. The diversity of p63 isoforms expressed in distinct tissues allows this transcription factor to have a wide array of effects. p63 coordinates the transcription of genes involved in cell survival, stem cell self-renewal, migration, differentiation, and epithelial-to-mesenchymal transition. Through the regulation of these biological processes, p63 contributes to, not only normal epithelial development, but also epithelium-derived cancer pathogenesis. In this review, we provide an overview of the role of p63 in epithelial stemness regulation, including self-renewal, differentiation, proliferation, and senescence. We describe the differential expression of TAp63 and ΔNp63 isoforms and their distinct functional activities in normal epithelial tissues and in epithelium-derived tumors. Furthermore, we summarize the signaling cascades modulating the TAp63 and ΔNp63 isoforms as well as their downstream pathways in stemness regulation.


Subject(s)
Neoplasms , Tumor Suppressor Proteins , Humans , Tumor Suppressor Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Epithelium/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Protein Isoforms/metabolism , Phosphoproteins/genetics
6.
Cell Death Dis ; 14(10): 691, 2023 10 20.
Article in English | MEDLINE | ID: mdl-37863894

ABSTRACT

Atherosclerosis is a chronic inflammatory disease characterized by the accumulation of fatty deposits in the inner walls of vessels. These plaques restrict blood flow and lead to complications such as heart attack or stroke. The development of atherosclerosis is influenced by a variety of factors, including age, genetics, lifestyle, and underlying health conditions such as high blood pressure or diabetes. Atherosclerotic plaques in stable form are characterized by slow growth, which leads to luminal stenosis, with low embolic potential or in unstable form, which contributes to high risk for thrombotic and embolic complications with rapid clinical onset. In this complex scenario of atherosclerosis, macrophages participate in the whole process, including the initiation, growth and eventually rupture and wound healing stages of artery plaque formation. Macrophages in plaques exhibit high heterogeneity and plasticity, which affect the evolving plaque microenvironment, e.g., leading to excessive lipid accumulation, cytokine hyperactivation, hypoxia, apoptosis and necroptosis. The metabolic and functional transitions of plaque macrophages in response to plaque microenvironmental factors not only influence ongoing and imminent inflammatory responses within the lesions but also directly dictate atherosclerotic progression or regression. In this review, we discuss the origin of macrophages within plaques, their phenotypic diversity, metabolic shifts, and fate and the roles they play in the dynamic progression of atherosclerosis. It also describes how macrophages interact with other plaque cells, particularly T cells. Ultimately, targeting pathways involved in macrophage polarization may lead to innovative and promising approaches for precision medicine. Further insights into the landscape and biological features of macrophages within atherosclerotic plaques may offer valuable information for optimizing future clinical treatment for atherosclerosis by targeting macrophages.


Subject(s)
Atherosclerosis , Myocardial Infarction , Plaque, Atherosclerotic , Humans , Plaque, Atherosclerotic/pathology , Atherosclerosis/pathology , Macrophages/metabolism , Apoptosis , Myocardial Infarction/metabolism
7.
Oncogene ; 42(44): 3221-3235, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37704784

ABSTRACT

Chemotherapy resistance represents a major cause of therapeutic failure and mortality in cancer patients. Mesenchymal stromal cells (MSCs), an integral component of tumor microenvironment, are known to promote drug resistance. However, the detailed mechanisms remain to be elucidated. Here, we found that MSCs confer breast cancer resistance to doxorubicin by diminishing its intratumoral accumulation. Hyaluronan (HA), a major extracellular matrix (ECM) product of MSCs, was found to mediate the chemoresistant effect. The chemoresistant effect of MSCs was abrogated when hyaluronic acid synthase 2 (HAS2) was depleted or inhibited. Exogenous HA also protected tumor grafts from doxorubicin. Molecular dynamics simulation analysis indicates that HA can bind with doxorubicin, mainly via hydrophobic and hydrogen bonds, and thus reduce its entry into breast cancer cells. This mechanism is distinct from the reported chemoresistant effect of HA via its receptor on cell surface. High HA serum levels were also found to be positively associated with chemoresistance in breast cancer patients. Our findings indicate that the HA-doxorubicin binding dynamics can confer cancer cells chemoresistance. Reducing HA may enhance chemotherapy efficacy.


Subject(s)
Breast Neoplasms , Mesenchymal Stem Cells , Humans , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Hyaluronic Acid/metabolism , Doxorubicin/pharmacology , Hyaluronan Synthases/metabolism , Extracellular Matrix/metabolism , Mesenchymal Stem Cells/metabolism , Hyaluronan Receptors/metabolism , Tumor Microenvironment
8.
Cell Mol Immunol ; 20(10): 1171-1185, 2023 10.
Article in English | MEDLINE | ID: mdl-37580400

ABSTRACT

Mesenchymal stem/stromal cells (MSCs) possess robust immunoregulatory functions and are promising therapeutics for inflammatory disorders. This capacity is not innate but is activated or 'licensed' by inflammatory cytokines. The licensing mechanism remains unclear. Here, we examined whether inflammatory cytokines metabolically reprogrammed MSCs to confer this immunoregulatory capacity. In response to stimulation by inflammatory cytokines, MSCs exhibited a dramatic increase in the consumption of glucose, which was accompanied by an enhanced use of nicotinamide adenine dinucleotide (NAD+) and increased expression of nicotinamide phosphoribosyltransferase (NAMPT), a central enzyme in the salvage pathway for NAD+ production. When NAD+ synthesis was blocked by inhibiting or depleting NAMPT, the immunosuppressive function of MSCs induced by inflammatory cytokines was greatly attenuated. Consequently, when NAD+ metabolism in MSCs was perturbed, their therapeutic benefit was decreased in mice suffering from inflammatory bowel disease and acute liver injury. Further analysis revealed that NAMPT-driven production of NAD+ was critical for the inflammatory cytokine-induced increase in glycolysis in MSCs. Furthermore, the increase in glycolysis led to succinate accumulation in the tricarboxylic acid cycle, which led to hypoxia-inducible factor 1α (HIF-1α) stabilization and subsequently increased the transcription of key glycolytic genes, thereby persistently maintaining glycolytic flux. This study demonstrated that unlike its proinflammatory role in immune cells, NAD+ metabolism governs the anti-inflammatory function of MSCs during inflammation.


Subject(s)
Mesenchymal Stem Cells , NAD , Mice , Animals , NAD/metabolism , Glycolysis , Citric Acid Cycle , Cytokines/metabolism , Mesenchymal Stem Cells/metabolism
9.
Cell Death Discov ; 9(1): 269, 2023 Jul 28.
Article in English | MEDLINE | ID: mdl-37507432

ABSTRACT

Muscle stem cells (MuSCs) have been demonstrated to exert impressive therapeutic efficacy in disease settings through orchestrating inflammatory microenvironments. Nevertheless, the mechanisms underlying the immunoregulatory property of MuSCs remain largely uncharacterized. Here, we showed that interleukin-4-induced-1 (IL4I1), an essential enzyme that catalyzes indole metabolism in humans, was highly expressed in human MuSCs exposed to IFN-γ and TNF-α. Functionally, the MuSCs were found to inhibit the infiltration of neutrophils into sites of inflammation in a IL4I1-dependent manner and thus ameliorate acute lung injury in mice. Mechanistically, the indole metabolites, including indole-3-pyruvic acid (I3P) and indole-3-aldehyde (I3A), produced by IL4I1, acted as ligands to activate aryl hydrocarbon receptor (AHR), leading to augmented expression of TNF-stimulated gene 6 (TSG-6) in inflammatory cytokine-primed MuSCs. Furthermore, I3P administration alone suppressed neutrophil infiltration into damaged lungs. I3P could also reduce the level of reactive oxygen species in neutrophils. Therefore, our study has uncovered a novel mechanism by which MuSCs acquire their immunoregulatory property and may help to develop or optimize MuSC-based therapies for inflammatory diseases.

10.
Cell Biosci ; 13(1): 81, 2023 May 10.
Article in English | MEDLINE | ID: mdl-37165408

ABSTRACT

Nicotinamide adenine dinucleotide (NAD+) is a critical metabolite that acts as a cofactor in energy metabolism, and serves as a cosubstrate for non-redox NAD+-dependent enzymes, including sirtuins, CD38 and poly(ADP-ribose) polymerases. NAD+ metabolism can regulate functionality attributes of innate and adaptive immune cells and contribute to inflammatory responses. Thus, the manipulation of NAD+ bioavailability can reshape the courses of immunological diseases. Here, we review the basics of NAD+ biochemistry and its roles in the immune response, and discuss current challenges and the future translational potential of NAD+ research in the development of therapeutics for inflammatory diseases, such as COVID-19.

11.
Cell Stem Cell ; 29(11): 1515-1530, 2022 11 03.
Article in English | MEDLINE | ID: mdl-36332569

ABSTRACT

Mesenchymal stem/stromal cells (MSCs) exist in almost all tissues and participate in tissue regeneration and homeostasis. In-vitro-expanded MSCs are employed as therapeutics for autoimmune diseases, organ failures, and many other chronic disorders. Remarkably, the reparative and homeostatic maintenance functions of MSCs rely on their interaction with the inflammatory microenvironment. Here, we discuss the characteristics and functions of MSCs under different pathophysiological conditions and highlight how the immunomodulatory functions of MSCs are altered in accordance with the inflammatory cues. We hope to provide new insights into the diverse immunoregulatory properties of MSCs during tissue regeneration and therapy.


Subject(s)
Autoimmune Diseases , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Humans , Mesenchymal Stem Cells/physiology , Immunomodulation , Wound Healing , Immunity
12.
Cell Death Dis ; 13(11): 996, 2022 11 25.
Article in English | MEDLINE | ID: mdl-36433947

ABSTRACT

Psoriasis is currently an incurable skin disorder mainly driven by a chronic inflammatory response. We found that subcutaneous application of umbilical cord- derived mesenchymal stem/stromal cells (MSCs) primed by IFN-γ and TNF-α, referred to as MSCs-IT, exhibited remarkable therapeutic efficacy on imiquimod (IMQ)-induced psoriasis-like inflammation in mice. Neutrophil infiltration, a hallmark of psoriasis, was significantly reduced after treatment with MSCs-IT. We further demonstrated that the effects of MSCs-IT were mediated by tumor necrosis factor (TNF) stimulating gene-6 (TSG-6), which was greatly upregulated in MSCs upon IFN-γ and TNF-α stimulation. MSCs transduced with TSG-6 siRNA lost their therapeutic efficacy while recombinant TSG-6 applied alone could also reduce neutrophil infiltration and alleviate the psoriatic lesions. Furthermore, we demonstrated that TSG-6 could inhibit neutrophil recruitment by decreasing the expression of CXCL1, which may be related to the reduced level of STAT1 phosphorylation in the keratinocytes. Thus, blocking neutrophil recruitment by MSCs-IT or TSG-6 has potential for therapeutic application in human psoriasis.


Subject(s)
Mesenchymal Stem Cells , Neutrophils , Psoriasis , Animals , Humans , Mice , Cytokines , Immunologic Factors , Inflammation/genetics , Inflammation/immunology , Mesenchymal Stem Cells/immunology , Neutrophils/immunology , Psoriasis/genetics , Psoriasis/immunology , Psoriasis/therapy , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/immunology
13.
J Gastrointest Oncol ; 13(4): 1875-1888, 2022 Aug.
Article in English | MEDLINE | ID: mdl-36092323

ABSTRACT

Background: This study aimed to verify the hypothesis that circular RNA MMP9 (circMMP9) promotes hepatocellular carcinoma (HCC) progression through targeting miR-149 and regulating cyclin D2 (CCND2) expression. Methods: Expression of circMMP9, miR-149 and CCND2 was detected by quantitative reverse transcription polymerase chain reaction (qRT-PCR) or protein blotting. Cell Counting Kit-8 (CCK-8) was used to evaluate cell proliferation. The HCC cell migration and invasion were evaluated using wound healing and transwell assays. The interaction among circMMP9, miR-149, and CCND2 was evaluated using luciferase, RNA-pull down, and RNA immunoprecipitation (RIP) assays, respectively. Cell apoptosis and cycle were examined by flow cytometry. A subcutaneous HCC xenograft mouse model was established for analyzing the role of circMMP9 in regulating the progression of HCC in vivo. Results: The expression of circMMP9 was elevated in HCC tissues and its high expression correlated with poor prognosis (P<0.05). Knockdown of circMMP9 restrained the proliferation, migration, and invasion of HCC cells and led to arrested cell cycle and increased apoptosis (all P<0.05). Furthermore, knockdown of circMMP9 attenuated HCC growth in vivo (P<0.05). Mechanically, circMMP9 acted as a sponge for miR-149 and enhanced CCND2 expression in HCC cells (P<0.05). Inhibition of miR-149 or overexpression of CCND2 abrogated knockdown of circMMP9-mediated alleviation of the malignant phenotypes of HCC (P<0.05). Conclusions: For the first time, we demonstrated that circMMP9 exacerbated HCC progression through the miR-149/CCND2 axis, which suggested that circMMP9 could be potentially targeted for HCC treatment.

14.
Signal Transduct Target Ther ; 7(1): 92, 2022 03 21.
Article in English | MEDLINE | ID: mdl-35314676

ABSTRACT

Mesenchymal stromal/stem cells (MSCs) possess multi-lineage differentiation and self-renewal potentials. MSCs-based therapies have been widely utilized for the treatment of diverse inflammatory diseases, due to the potent immunoregulatory functions of MSCs. An increasing body of evidence indicates that MSCs exert their therapeutic effects largely through their paracrine actions. Growth factors, cytokines, chemokines, extracellular matrix components, and metabolic products were all found to be functional molecules of MSCs in various therapeutic paradigms. These secretory factors contribute to immune modulation, tissue remodeling, and cellular homeostasis during regeneration. In this review, we summarize and discuss recent advances in our understanding of the secretory behavior of MSCs and the intracellular communication that accounts for their potential in treating human diseases.


Subject(s)
Mesenchymal Stem Cells , Cell Differentiation/genetics , Cytokines/metabolism , Humans
15.
Front Oncol ; 12: 844182, 2022.
Article in English | MEDLINE | ID: mdl-35252013

ABSTRACT

BACKGROUND: Solid pseudopapillary neoplasms of the pancreas (SPNs) in male patients are more frequently reported. The aim of the study was to evaluate the sex features of SPN and the risk factors that predict tumor recurrence. METHODS: From 2013 to 2019, patients who were pathologically confirmed to have SPNs were retrospectively reviewed. The baseline study parameters were compared between males and females. A logistic regression model was established to identify the independent risk factors for tumor recurrence. RESULTS: In total, 221 patients were included in this study. Of them, 53 patients (24.0%) were males. Male patients were older than female patients (39.1 vs 31.6 years, P=0.001), and the tumor size in male patients was smaller than that in female patients (50.38 vs 39.65 mm, P=0.038). The preoperative imaging diagnostic accuracy was significantly higher in females than in males (70.5% vs 54%, P=0.02). SPNs in male patients tended to be misdiagnosed with other malignant tumors (37.7% vs 10.7%, P<0.0001), with a more solid component observed in images (66.8% vs 24.7%, P<0.0001). For immunohistochemical staining, the expression of beta catenin was significantly lower in male patients (P=0.002), and the expression of vimentin was the opposite (P=0.01). The overall survival rate and disease-free survival were not different. Based on multivariate analysis, older age [hazard ratio (HR)= 1.094, 95% confidence interval (CI): 1.005-1.190] and KI 67 index grade III (HR=12.029, 95% CI: 2.399-60.311) were independent risk factors for tumor recurrence. CONCLUSION: The clinical and imaging features of SPN in males were not in full accord with those in females; however, the differences did not influence prognosis.

16.
Biol Direct ; 16(1): 18, 2021 10 20.
Article in English | MEDLINE | ID: mdl-34670590

ABSTRACT

Skeletal muscle has an extraordinary regenerative capacity reflecting the rapid activation and effective differentiation of muscle stem cells (MuSCs). In the course of muscle regeneration, MuSCs are reprogrammed by immune cells. In turn, MuSCs confer immune cells anti-inflammatory properties to resolve inflammation and facilitate tissue repair. Indeed, MuSCs can exert therapeutic effects on various degenerative and inflammatory disorders based on their immunoregulatory ability, including effects primed by interferon-γ (IFN-γ) and tumor necrosis factor-α (TNF-α). At the molecular level, the tryptophan metabolites, kynurenine or kynurenic acid, produced by indoleamine 2,3-dioxygenase (IDO), augment the expression of TNF-stimulated gene 6 (TSG6) through the activation of the aryl hydrocarbon receptor (AHR). In addition, insulin growth factor 2 (IGF2) produced by MuSCs can endow maturing macrophages oxidative phosphorylation (OXPHOS)-dependent anti-inflammatory functions. Herein, we summarize the current understanding of the immunomodulatory characteristics of MuSCs and the issues related to their potential applications in pathological conditions, including COVID-19.


Subject(s)
COVID-19/therapy , Immune System/physiology , Muscles/physiology , Regeneration/physiology , Stem Cells/cytology , Animals , COVID-19/immunology , Cell Adhesion Molecules/metabolism , Cell Differentiation , Cell Proliferation , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Inflammation , Insulin-Like Growth Factor II/metabolism , Interferon-gamma/metabolism , Kynurenic Acid/metabolism , Kynurenine/metabolism , Macrophages/metabolism , Mice , Muscles/metabolism , Oxidative Phosphorylation , Receptors, Aryl Hydrocarbon/metabolism , Tryptophan/chemistry , Tumor Necrosis Factor-alpha/metabolism
17.
Oncogene ; 40(10): 1836-1850, 2021 03.
Article in English | MEDLINE | ID: mdl-33564072

ABSTRACT

Despite the widespread use of the blockade of immune checkpoints, for a significant number of cancer patients, these therapies have proven ineffective, presumably due to the immunosuppressive nature of the tumor microenvironment (TME). Critical drivers of immune escape in the TME include tumor-associated macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs), which not only mediate immune suppression, but also facilitate metastatic dissemination and impart resistance to immunotherapies. Thus, strategies that convert them into tumor fighters may offer great therapeutic potential. In this study, we evaluated whether pharmacologic modulation of macrophage phenotype by HDAC inhibitors (HDACi) could produce an anti-tumor effect. We demonstrated that low-dose HDACi trichostatin-A (TSA) markedly reshaped the tumor immune microenvironment by modulating the suppressive activity of infiltrating macrophages and inhibiting the recruitment of MDSCs in various tumors. These actions, in turn, augmented anti-tumor immune responses and further enhanced anti-tumor effects of immunotherapies. HDAC inhibition, however, also upregulated PD-L1, thereby limiting the beneficial therapeutic effects. Indeed, combining low-dose TSA with anti-PD-L1 in this model significantly enhanced the durability of tumor reduction and prolonged survival of tumor-bearing mice, compared with the effect of either treatment alone. These data introduce HDAC inhibition as a potential means to harness the anti-tumor potential of macrophages in cancer therapy.


Subject(s)
B7-H1 Antigen/genetics , Histone Deacetylases/genetics , Hydroxamic Acids/pharmacology , Melanoma, Experimental/drug therapy , Animals , B7-H1 Antigen/antagonists & inhibitors , Disease Models, Animal , Heterografts , Histone Deacetylases/drug effects , Humans , Immune Checkpoint Inhibitors/pharmacology , Melanoma, Experimental/genetics , Melanoma, Experimental/pathology , Mice , Myeloid-Derived Suppressor Cells/drug effects , Tumor Microenvironment/drug effects , Tumor-Associated Macrophages/drug effects
18.
Stem Cell Res Ther ; 12(1): 50, 2021 01 09.
Article in English | MEDLINE | ID: mdl-33422134

ABSTRACT

BACKGROUND: Muscle stem cells (MuSCs) are absolutely required for the formation, repair, and regeneration of skeletal muscle tissue. Increasing evidence demonstrated that tissue stem cells, especially mesenchymal stem cells (MSCs), can exert therapeutic effects on various degenerative and inflammatory disorders based on their immunoregulatory properties. Human mesenchymal stem cells (hMSCs) treated with interferon-γ (IFN-γ) and tumor necrosis factor-α (TNF-α) were reported to possess anti-inflammatory functions by producing TNF-stimulated gene 6 (TSG-6). However, whether human muscle stem cells (hMuSCs) also possess TSG-6 mediated anti-inflammatory functions has not been explored. METHODS: The ulcerative colitis mouse model was established by subjecting mice to dextran sulfate sodium (DSS) in drinking water for 7 days. hMuSCs were pretreated with IFN-γ and TNF-α for 48 h and were then transplanted intravenously at day 2 of DSS administration. Body weights were monitored daily. Indoleamine 2,3-dioxygenase (IDO) and TSG-6 in hMuSCs were knocked down with short hairpin RNA (shRNA) and small interfering RNA (siRNA), respectively. Colon tissues were collected for length measurement and histopathological examination. The serum level of IL-6 in mice was measured by enzyme-linked immunosorbent assay (ELISA). Real-time PCR and Western blot analysis were performed to evaluate gene expression. RESULTS: hMuSCs treated with inflammatory factors significantly ameliorated inflammatory bowel disease (IBD) symptoms. IDO and TSG-6 were greatly upregulated and required for the beneficial effects of hMuSCs on IBD. Mechanistically, the tryptophan metabolites, kynurenine (KYN) or kynurenic acid (KYNA) produced by IDO, augmented the expression of TSG-6 through activating their common receptor aryl hydrocarbon receptor (AHR). CONCLUSION: Inflammatory cytokines-treated hMuSCs can alleviate DSS-induced colitis through IDO-mediated TSG-6 production.


Subject(s)
Colitis, Ulcerative , Colitis , Mesenchymal Stem Cells , Animals , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/therapy , Cytokines , Dextran Sulfate , Humans , Mice , Muscles
19.
Free Radic Biol Med ; 161: 339-350, 2020 12.
Article in English | MEDLINE | ID: mdl-33122005

ABSTRACT

Distinct metabolic programs, either energy-consuming anabolism or energy-generating catabolism, were required for different biological functions. Macrophages can adopt different immune phenotypes in response to various cues and exhibit anti- or pro-inflammatory properties relying on catabolic pathways associated with oxidative phosphorylation (OXPHOS) or glycolysis. Spermidine, a natural polyamine, has been reported to regulate inflammation through inducing anti-inflammatory (M2) macrophages. However, the underlying mechanisms remain elusive. We show here that the M2-polarization induced by spermidine is mediated by mitochondrial reactive oxygen species (mtROS). The levels of mitochondrial superoxide and H2O2 were markedly elevated by spermidine. Mechanistically, mtROS were found to activate AMP-activated protein kinase (AMPK), which in turn enhanced mitochondrial function. Furthermore, hypoxia-inducible factor-1α (Hif-1α) was upregulated by the AMPK activation and mtROS and was required for the expression of anti-inflammatory genes and induction of autophagy. Consistent with previous report that autophagy is required for the M2 polarization, we found that the M2 polarization induced by spermidine was also mediated by increased autophagy. The macrophages treated with spermidine in vitro were found to ameliorate Dextran Sulfate Sodium (DSS)-induced inflammatory bowel disease (IBD) in mice. Thus, spermidine can elicit an anti-inflammatory program driven by mtROS-dependent AMPK activation, Hif-1α stabilization and autophagy induction in macrophages. Our studies revealed a critical role of mtROS in shaping macrophages into M2-like phenotype and provided novel information for management of inflammatory disease by spermidine.


Subject(s)
AMP-Activated Protein Kinases , Spermidine , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Autophagy , Hydrogen Peroxide/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Macrophages/metabolism , Mice , Mitochondria/metabolism , Spermidine/metabolism , Spermidine/pharmacology , Superoxides/metabolism , Up-Regulation
20.
Cell Death Dis ; 11(8): 707, 2020 08 26.
Article in English | MEDLINE | ID: mdl-32848141

ABSTRACT

The dome-shaped cornea is a transparent, non-vascularized, and epithelialized highly organized tissue. Physical and chemical injuries may trigger corneal wound healing (CWH) response and result in neovascularization that impairs the visual function. CWH involves not only migration, proliferation, and differentiation of the cells in different layers of cornea, but also the mobilization of immune cells. We demonstrated here that human adipose-derived mesenchymal stromal cells (ADSCs) could effectively inhibit neovascularization during ethanol-induced injury in mouse cornea. Importantly, we found that while neutrophils are essential for CWH, excessive and prolonged neutrophil retention during the granulation stage contributes to neovascularization. ADSCs were found to promote the clearance of neutrophils in the cornea during the granulation stage, likely via increasing the reverse transendothelial cell migration of CXCR4high neutrophils from cornea to the lung. Our results demonstrate that ADSCs are effective in treating CWH-induced neovascularization and modulation of neutrophil clearance could be novel strategies for better vision recovery after injury.


Subject(s)
Cornea/metabolism , Mesenchymal Stem Cells/metabolism , Wound Healing/physiology , Animals , Cell Differentiation , China , Corneal Injuries/metabolism , Corneal Injuries/therapy , Female , Humans , Male , Mesenchymal Stem Cell Transplantation/methods , Mice , Mice, Inbred BALB C , Neutrophils/metabolism , Skin/cytology , Wound Healing/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...