Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.787
Filter
1.
Nat Commun ; 15(1): 5731, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38977708

ABSTRACT

Neuropilin-1 (NRP1), a co-receptor for various cytokines, including TGF-ß, has been identified as a potential therapeutic target for fibrosis. However, its role and mechanism in renal fibrosis remains elusive. Here, we show that NRP1 is upregulated in distal tubular (DT) cells of patients with transplant renal insufficiency and mice with renal ischemia-reperfusion (I-R) injury. Knockout of Nrp1 reduces multiple endpoints of renal injury and fibrosis. We find that Nrp1 facilitates the binding of TNF-α to its receptor in DT cells after renal injury. This signaling results in a downregulation of lysine crotonylation of the metabolic enzyme Cox4i1, decreases cellular energetics and exacerbation of renal injury. Furthermore, by single-cell RNA-sequencing we find that Nrp1-positive DT cells secrete collagen and communicate with myofibroblasts, exacerbating acute kidney injury (AKI)-induced renal fibrosis by activating Smad3. Dual genetic deletion of Nrp1 and Tgfbr1 in DT cells better improves renal injury and fibrosis than either single knockout. Together, these results reveal that targeting of NRP1 represents a promising strategy for the treatment of AKI and subsequent chronic kidney disease.


Subject(s)
Acute Kidney Injury , Fibrosis , Mice, Knockout , Neuropilin-1 , Receptor, Transforming Growth Factor-beta Type I , Reperfusion Injury , Smad3 Protein , Neuropilin-1/metabolism , Neuropilin-1/genetics , Animals , Humans , Mice , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Acute Kidney Injury/genetics , Receptor, Transforming Growth Factor-beta Type I/metabolism , Receptor, Transforming Growth Factor-beta Type I/genetics , Reperfusion Injury/metabolism , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Smad3 Protein/metabolism , Smad3 Protein/genetics , Male , Tumor Necrosis Factor-alpha/metabolism , Signal Transduction , Mice, Inbred C57BL , Kidney Tubules/pathology , Kidney Tubules/metabolism , Myofibroblasts/metabolism , Myofibroblasts/pathology , Collagen/metabolism
2.
J Transl Med ; 22(1): 617, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38961399

ABSTRACT

INTRODUCTION: Intrauterine adhesions (IUA) manifest as endometrial fibrosis, often causing infertility or recurrent miscarriage; however, their pathogenesis remains unclear. OBJECTIVES: This study assessed the role of Dickkopf WNT signaling pathway inhibitor 1 (DKK1) and autophagy in endometrial fibrosis, using clinical samples as well as in vitro and in vivo experiments. METHODS: Immunohistochemistry, immunofluorescence and western blot were used to determine the localization and expression of DKK1 in endometrium; DKK1 silencing and DKK1 overexpression were used to detect the biological effects of DKK1 silencing or expression in endometrial cells; DKK1 gene knockout mice were used to observe the phenotypes caused by DKK1 gene knockout. RESULTS: In patients with IUA, DKK1 and autophagy markers were down-regulated; also, α-SMA and macrophage localization were increased in the endometrium. DKK1 conditional knockout (CKO) mice showed a fibrotic phenotype with decreased autophagy and increased localization of α-SMA and macrophages in the endometrium. In vitro studies showed that DKK1 knockout (KO) suppressed the autophagic flux of endometrial stromal cells. In contrast, ectopic expression of DKK1 showed the opposite phenotype. Mechanistically, we discovered that DKK1 regulates autophagic flux through Wnt/ß-catenin and PI3K/AKT/mTOR pathways. Further studies showed that DKK1 KO promoted the secretion of interleukin (IL)-8 in exosomes, thereby promoting macrophage proliferation and metastasis. Also, in DKK1 CKO mice, treatment with autophagy activator rapamycin partially restored the endometrial fibrosis phenotype. CONCLUSION: Our findings indicated that DKK1 was a potential diagnostic marker or therapeutic target for IUA.


Subject(s)
Autophagy , Endometrium , Exosomes , Fibrosis , Intercellular Signaling Peptides and Proteins , Macrophages , Mice, Knockout , Myofibroblasts , Animals , Female , Intercellular Signaling Peptides and Proteins/metabolism , Endometrium/metabolism , Endometrium/pathology , Macrophages/metabolism , Macrophages/pathology , Humans , Exosomes/metabolism , Myofibroblasts/metabolism , Myofibroblasts/pathology , Mice , Mice, Inbred C57BL , Adult
3.
Nat Commun ; 15(1): 5895, 2024 Jul 13.
Article in English | MEDLINE | ID: mdl-39003267

ABSTRACT

Autoimmune thyroid diseases (AITD) such as Graves' disease (GD) or Hashimoto's thyroiditis (HT) are organ-specific diseases that involve complex interactions between distinct components of thyroid tissue. Here, we use spatial transcriptomics to explore the molecular architecture, heterogeneity and location of different cells present in the thyroid tissue, including thyroid follicular cells (TFCs), stromal cells such as fibroblasts, endothelial cells, and thyroid infiltrating lymphocytes. We identify damaged antigen-presenting TFCs with upregulated CD74 and MIF expression in thyroid samples from AITD patients. Furthermore, we discern two main fibroblast subpopulations in the connective tissue including ADIRF+ myofibroblasts, mainly enriched in GD, and inflammatory fibroblasts, enriched in HT patients. We also demonstrate an increase of fenestrated PLVAP+ vessels in AITD, especially in GD. Our data unveil stromal and thyroid epithelial cell subpopulations that could play a role in the pathogenesis of AITD.


Subject(s)
Antigens, Differentiation, B-Lymphocyte , Graves Disease , Hashimoto Disease , Thyroid Gland , Humans , Graves Disease/pathology , Graves Disease/immunology , Graves Disease/genetics , Graves Disease/metabolism , Thyroid Gland/pathology , Thyroid Gland/metabolism , Hashimoto Disease/pathology , Hashimoto Disease/immunology , Hashimoto Disease/metabolism , Hashimoto Disease/genetics , Antigens, Differentiation, B-Lymphocyte/metabolism , Antigens, Differentiation, B-Lymphocyte/genetics , Fibroblasts/metabolism , Fibroblasts/pathology , Histocompatibility Antigens Class II/metabolism , Histocompatibility Antigens Class II/genetics , Thyroid Epithelial Cells/metabolism , Thyroid Epithelial Cells/pathology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Transcriptome , Myofibroblasts/metabolism , Myofibroblasts/pathology , Stromal Cells/metabolism , Stromal Cells/pathology , Female , Macrophage Migration-Inhibitory Factors , Intramolecular Oxidoreductases
4.
Respir Res ; 25(1): 284, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39026235

ABSTRACT

Idiopathic pulmonary fibrosis is a lethal, progressive, and irreversible condition that has become a significant focus of medical research due to its increasing incidence. This rising trend presents substantial challenges for patients, healthcare providers, and researchers. Despite the escalating burden of pulmonary fibrosis, the available therapeutic options remain limited. Currently, the United States Food and Drug Administration has approved two drugs for the treatment of pulmonary fibrosis-nintedanib and pirfenidone. However, their therapeutic effectiveness is limited, and they cannot reverse the fibrosis process. Additionally, these drugs are associated with significant side effects. Myofibroblasts play a central role in the pathophysiology of pulmonary fibrosis, significantly contributing to its progression. Consequently, strategies aimed at inhibiting myofibroblast differentiation or promoting their dedifferentiation hold promise as effective treatments. This review examines the regulation of myofibroblast dedifferentiation, exploring various signaling pathways, regulatory targets, and potential pharmaceutical interventions that could provide new directions for therapeutic development.


Subject(s)
Cell Dedifferentiation , Myofibroblasts , Humans , Myofibroblasts/pathology , Myofibroblasts/metabolism , Myofibroblasts/drug effects , Cell Dedifferentiation/drug effects , Cell Dedifferentiation/physiology , Animals , Idiopathic Pulmonary Fibrosis/pathology , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/drug therapy , Signal Transduction/physiology , Antifibrotic Agents/therapeutic use , Antifibrotic Agents/pharmacology , Pulmonary Fibrosis/pathology , Pulmonary Fibrosis/metabolism
5.
Cell Death Dis ; 15(7): 493, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38987529

ABSTRACT

Lung cancer is a leading cause of cancer-related mortality globally, with a dismal 5-year survival rate, particularly for Lung Adenocarcinoma (LUAD). Mechanical changes within the tumor microenvironment, such as extracellular matrix (ECM) remodeling and fibroblast activity, play pivotal roles in cancer progression and metastasis. However, the specific impact of the basement membrane (BM) on the mechanical characteristics of LUAD remains unclear. This study aims to identify BM genes influencing internal mechanical stress in tumors, elucidating their effects on LUAD metastasis and therapy resistance, and exploring strategies to counteract these effects. Using Matrigel overlay and Transwell assays, we found that mechanical stress, mimicked by matrix application, augmented LUAD cell migration and invasion, correlating with ECM alterations and activation of the epithelial-mesenchymal transition (EMT) pathway. Employing machine learning, we developed the SVM_Score model based on relevant BM genes, which accurately predicted LUAD patient prognosis and EMT propensity across multiple datasets. Lower SVM_Scores were associated with worse survival outcomes, elevated cancer-related pathways, increased Tumor Mutation Burden, and higher internal mechanical stress in LUAD tissues. Notably, the SVM_Score was closely linked to COL5A1 expression in myofibroblasts, a key marker of mechanical stress. High COL5A1 expression from myofibroblasts promoted tumor invasiveness and EMT pathway activation in LUAD cells. Additionally, treatment with Sorafenib, which targets COL5A1 secretion, attenuated the tumor-promoting effects of myofibroblast-derived COL5A1, inhibiting LUAD cell proliferation, migration, and enhancing chemosensitivity. In conclusion, this study elucidates the complex interplay between mechanical stress, ECM alterations, and LUAD progression. The SVM_Score emerges as a robust prognostic tool reflecting tumor mechanical characteristics, while Sorafenib intervention targeting COL5A1 secretion presents a promising therapeutic strategy to mitigate LUAD aggressiveness. These findings deepen our understanding of the biomechanical aspects of LUAD and offer insights for future research and clinical applications.


Subject(s)
Adenocarcinoma of Lung , Collagen Type V , Epithelial-Mesenchymal Transition , Lung Neoplasms , Myofibroblasts , Stress, Mechanical , Humans , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/drug therapy , Myofibroblasts/metabolism , Myofibroblasts/drug effects , Myofibroblasts/pathology , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/drug therapy , Epithelial-Mesenchymal Transition/drug effects , Collagen Type V/metabolism , Collagen Type V/genetics , Drug Resistance, Neoplasm/drug effects , Cell Line, Tumor , Animals , Cell Movement/drug effects , Neoplasm Metastasis , Mice , Tumor Microenvironment , Sorafenib/pharmacology , Sorafenib/therapeutic use , Extracellular Matrix/metabolism
6.
Ann Clin Lab Sci ; 54(3): 363-370, 2024 May.
Article in English | MEDLINE | ID: mdl-39048163

ABSTRACT

OBJECTIVE: During the progression of chronic idiopathic pulmonary fibrosis (IPF), maladaptive tissue remodeling including excessive extracellular matrix (ECM) deposition occurs, which eventually leads to architectural distortion and loss of organ function in organ fibrosis. ADAM15, which is highly expressed in the developing lungs and kidneys, is a transmembrane-anchored multidomain protein belonging to the family of metalloproteinases. Compared to the extensive studies about functions of matrix metalloproteinases (MMPs), less are discussed about ADAM15, particularly in function and mechanism involving fibrogenesis. Our study aims to fill in this gap. METHODS: We identified ADAM15 as a novel antifibrotic mediator in lung fibrosis. We found that ADAM15 has cross-talks with transforming growth factor-ß1 (TGF-ß1), which is the most potent profibrotic mediator. We provided molecular and translational evidence that knockdown of ADAM15 accelerated fibrogenic response induced by TGF-ß1 and upregulation of ADAM15 rescued TGF-ß1-induced myofibroblast activation in part. RESULTS: Overexpression of ADAM15 ameliorates fibrotic changes and ADAM15 deficiency exacerbates changes from fibroblast to myofibroblast in NIH/3T3. Results were also presented and identified by the intuitive immunofluorescence staining. CONCLUSION: In this study, we uncover a new molecular mechanism of tissue fibrogenesis and identify ADAM15 as a potential therapeutic target in the treatment of fibrotic diseases.


Subject(s)
ADAM Proteins , Extracellular Matrix , Fibroblasts , Membrane Proteins , Transforming Growth Factor beta1 , Transforming Growth Factor beta1/metabolism , Animals , Mice , Fibroblasts/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , ADAM Proteins/metabolism , ADAM Proteins/genetics , Extracellular Matrix/metabolism , Humans , NIH 3T3 Cells , Myofibroblasts/metabolism , Myofibroblasts/pathology , Idiopathic Pulmonary Fibrosis/pathology , Idiopathic Pulmonary Fibrosis/metabolism
7.
ACS Appl Mater Interfaces ; 16(29): 37530-37544, 2024 Jul 24.
Article in English | MEDLINE | ID: mdl-38989714

ABSTRACT

Contrary to the initial belief that myofibroblasts are terminally differentiated cells, myofibroblasts have now been widely recognized as an activation state that is reversible. Therefore, strategies targeting myofibroblast to be a quiescent state may be an effective way for antihypertrophic scar therapy. Graphene quantum dots (GQDs), a novel zero-dimensional and carbon-based nanomaterial, have recently garnered significant interest in nanobiomedicine, owing to their excellent biocompatibility, tunable photoluminescence, and superior physiological stability. Although multiple nanoparticles have been used to alleviate hypertrophic scars, a GQD-based therapy has not been reported. Our in vivo studies showed that GQDs exhibited significant antiscar efficacy, with scar appearance improvement, collagen reduction and rearrangement, and inhibition of myofibroblast overproliferation. Further in vitro experiments revealed that GQDs inhibited α-SMA expression, collagen synthesis, and cell proliferation and migration, inducing myofibroblasts to become quiescent fibroblasts. Mechanistic studies have demonstrated that the effect of GQDs on myofibroblast proliferation blocked cell cycle progression by disrupting the cyclin-CDK-E2F axis. This study suggests that GQDs, which promote myofibroblast-to-fibroblast transition, could be a novel antiscar nanomedicine for the treatment of hypertrophic scars and other types of pathological fibrosis.


Subject(s)
Cell Proliferation , Cicatrix, Hypertrophic , Graphite , Myofibroblasts , Quantum Dots , Quantum Dots/chemistry , Myofibroblasts/drug effects , Myofibroblasts/pathology , Myofibroblasts/metabolism , Graphite/chemistry , Graphite/pharmacology , Cicatrix, Hypertrophic/drug therapy , Cicatrix, Hypertrophic/pathology , Cell Proliferation/drug effects , Animals , Humans , Mice , Collagen/chemistry , Cell Movement/drug effects
8.
Int J Mol Sci ; 25(14)2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39063116

ABSTRACT

The burden of chronic liver disease is globally increasing at an alarming rate. Chronic liver injury leads to liver inflammation and fibrosis (LF) as critical determinants of long-term outcomes such as cirrhosis, liver cancer, and mortality. LF is a wound-healing process characterized by excessive deposition of extracellular matrix (ECM) proteins due to the activation of hepatic stellate cells (HSCs). In the healthy liver, quiescent HSCs metabolize and store retinoids. Upon fibrogenic activation, quiescent HSCs transdifferentiate into myofibroblasts; lose their vitamin A; upregulate α-smooth muscle actin; and produce proinflammatory soluble mediators, collagens, and inhibitors of ECM degradation. Activated HSCs are the main effector cells during hepatic fibrogenesis. In addition, the accumulation and activation of profibrogenic macrophages in response to hepatocyte death play a critical role in the initiation of HSC activation and survival. The main source of myofibroblasts is resident HSCs. Activated HSCs migrate to the site of active fibrogenesis to initiate the formation of a fibrous scar. Single-cell technologies revealed that quiescent HSCs are highly homogenous, while activated HSCs/myofibroblasts are much more heterogeneous. The complex process of inflammation results from the response of various hepatic cells to hepatocellular death and inflammatory signals related to intrahepatic injury pathways or extrahepatic mediators. Inflammatory processes modulate fibrogenesis by activating HSCs and, in turn, drive immune mechanisms via cytokines and chemokines. Increasing evidence also suggests that cellular stress responses contribute to fibrogenesis. Recent data demonstrated that LF can revert even at advanced stages of cirrhosis if the underlying cause is eliminated, which inhibits the inflammatory and profibrogenic cells. However, despite numerous clinical studies on plausible drug candidates, an approved antifibrotic therapy still remains elusive. This state-of-the-art review presents cellular and molecular mechanisms involved in hepatic fibrogenesis and its resolution, as well as comprehensively discusses the drivers linking liver injury to chronic liver inflammation and LF.


Subject(s)
Hepatic Stellate Cells , Liver Cirrhosis , Humans , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Animals , Myofibroblasts/metabolism , Myofibroblasts/pathology
9.
Arkh Patol ; 86(4): 58-63, 2024.
Article in Russian | MEDLINE | ID: mdl-39073544

ABSTRACT

A literature review reflects data on the mechanisms of pulmonary fibrosis after a novel coronavirus infection associated with the SARS-COV2 virus. Factors contributing to post-COVID lung remodeling are considered. According to the literature, in the mechanism of pulmonary fibrosis, during the course of the disease and during the recovery period, both direct viral damage and death of alveolocytes and endothelium, the development of a systemic inflammatory reaction due to inadequate secretion of cytokines, especially type 2, which are activators of the proliferation of fibroblasts and myofibroblasts, are important. The influence of angiogenesis disorders and vascular dysfunction on pneumofibrosis was noted. Attention is also paid to the relationship between the development of pulmonary fibrosis and abnormal activation of the renin-angiotensin-aldosterone system. In combination with the action of many factors, especially germinal ones, an imbalance between profibrogenic and antifibrogenic action develops and fibrosis occurs.


Subject(s)
COVID-19 , Pulmonary Fibrosis , SARS-CoV-2 , Humans , Pulmonary Fibrosis/pathology , Pulmonary Fibrosis/etiology , Pulmonary Fibrosis/metabolism , COVID-19/complications , COVID-19/pathology , Renin-Angiotensin System , Cytokines/metabolism , Fibroblasts/pathology , Fibroblasts/metabolism , Myofibroblasts/pathology , Myofibroblasts/metabolism
10.
Iran J Allergy Asthma Immunol ; 23(2): 197-220, 2024 Apr 07.
Article in English | MEDLINE | ID: mdl-38822514

ABSTRACT

Systemic sclerosis (SSc) is an autoimmune systemic disease that is characterized by immune dysregulation, inflammation, vasculopathy, and fibrosis. Tissue fibrosis plays an important role in SSc and can affect several organs such as the dermis, lungs, and heart. Dysregulation of interferon (IFN) signaling contributes to the SSc pathogenesis and interferon regulatory factor 1 (IRF1) has been indicated as the main regulator of type I IFN. This study aimed to clarify the effect of IFN-gamma (-γ) and dexamethasone (DEX) on the IRF1, extracellular signal-regulated kinase 1/2 (ERK1/2), and the expression of alpha-smooth muscle actin (α-SMA) in myofibroblasts and genes involved in the inflammation and fibrosis processes in early diffuse cutaneous systemic sclerosis (dcSSc). A total of 10 early dcSSc patients (diffuse cutaneous form) and 10 unaffected control dermis biopsies were obtained to determine IFNγ and DEX effects on inflammation and fibrosis. Fibroblasts were treated with IFNγ and DEX at optimum time and dose. The expression level of genes and proteins involved in the fibrosis and inflammation processes have been quantified by quantitative real-time PCR (RT-qPCR) and western blot, respectively. IFNγ could up-regulate some of the inflammation-related genes (Interleukin-6; IL6) and down-regulate some of the fibrosis-related genes (COL1A1) in cultured fibroblasts of patients with early dcSSc compared to the untreated group. Besides, it has been revealed that IFNγ can induce fibroblast differentiation to the myofibroblast that expresses α-SMA. Concerning the inhibitory effect of IFNγ on some fibrotic genes and its positive effect on the inflammatory genes and myofibroblast differentiation, it seems that IFNγ may play a dual role in SSc.


Subject(s)
Actins , Fibroblasts , Interferon-gamma , Interleukin-6 , Scleroderma, Systemic , Adult , Female , Humans , Male , Middle Aged , Actins/metabolism , Actins/genetics , Cells, Cultured , Dexamethasone/pharmacology , Fibroblasts/metabolism , Fibroblasts/pathology , Fibroblasts/drug effects , Fibrosis , Gene Expression Regulation/drug effects , Interferon Regulatory Factor-1/metabolism , Interferon Regulatory Factor-1/genetics , Interferon-gamma/pharmacology , Interleukin-6/metabolism , Interleukin-6/genetics , Myofibroblasts/metabolism , Myofibroblasts/pathology , Scleroderma, Systemic/metabolism , Scleroderma, Systemic/pathology , Scleroderma, Systemic/immunology
12.
J Transl Med ; 22(1): 561, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38867256

ABSTRACT

BACKGROUND: Fibrogenesis within ovarian endometrioma (endometrioma), mainly induced by transforming growth factor-ß (TGF-ß), is characterized by myofibroblast over-activation and excessive extracellular matrix (ECM) deposition, contributing to endometrioma-associated symptoms such as infertility by impairing ovarian reserve and oocyte quality. However, the precise molecular mechanisms that underpin the endometrioma- associated fibrosis progression induced by TGF-ß remain poorly understood. METHODS: The expression level of lysine acetyltransferase 14 (KAT14) was validated in endometrium biopsies from patients with endometrioma and healthy controls, and the transcription level of KAT14 was further confirmed by analyzing a published single-cell transcriptome (scRNA-seq) dataset of endometriosis. We used overexpression, knockout, and knockdown approaches in immortalized human endometrial stromal cells (HESCs) or human primary ectopic endometrial stromal cells (EcESCs) to determine the role of KAT14 in TGF-ß-induced fibrosis. Furthermore, an adeno-associated virus (AAV) carrying KAT14-shRNA was used in an endometriosis mice model to assess the role of KAT14 in vivo. RESULTS: KAT14 was upregulated in ectopic lesions from endometrioma patients and predominantly expressed in activated fibroblasts. In vitro studies showed that KAT14 overexpression significantly promoted a TGF-ß-induced profibrotic response in endometrial stromal cells, while KAT14 silencing showed adverse effects that could be rescued by KAT14 re-enhancement. In vivo, Kat14 knockdown ameliorated fibrosis in the ectopic lesions of the endometriosis mouse model. Mechanistically, we showed that KAT14 directly interacted with serum response factor (SRF) to promote the expression of α-smooth muscle actin (α-SMA) by increasing histone H4 acetylation at promoter regions; this is necessary for TGF-ß-induced ECM production and myofibroblast differentiation. In addition, the knockdown or pharmacological inhibition of SRF significantly attenuated KAT14-mediating profibrotic effects under TGF-ß treatment. Notably, the KAT14/SRF complex was abundant in endometrioma samples and positively correlated with α-SMA expression, further supporting the key role of KAT14/SRF complex in the progression of endometrioma-associated fibrogenesis. CONCLUSION: Our results shed light on KAT14 as a key effector of TGF-ß-induced ECM production and myofibroblast differentiation in EcESCs by promoting histone H4 acetylation via co-operating with SRF, representing a potential therapeutic target for endometrioma-associated fibrosis.


Subject(s)
Endometriosis , Fibrosis , Serum Response Factor , Transforming Growth Factor beta , Adult , Animals , Female , Humans , Mice , Endometriosis/pathology , Endometriosis/metabolism , Endometrium/metabolism , Endometrium/pathology , Histone Acetyltransferases/metabolism , Myofibroblasts/metabolism , Myofibroblasts/pathology , Serum Response Factor/metabolism , Stromal Cells/metabolism , Stromal Cells/pathology , Transforming Growth Factor beta/metabolism , Up-Regulation/drug effects , Adaptor Proteins, Signal Transducing/metabolism
13.
Respir Res ; 25(1): 242, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38877465

ABSTRACT

BACKGROUND: Silicosis represents a paramount occupational health hazard globally, with its incidence, morbidity, and mortality on an upward trajectory, posing substantial clinical dilemmas due to limited effective treatment options available. Trigonelline (Trig), a plant alkaloid extracted mainly from coffee and fenugreek, have diverse biological properties such as protecting dermal fibroblasts against ultraviolet radiation and has the potential to inhibit collagen synthesis. However, it's unclear whether Trig inhibits fibroblast activation to attenuate silicosis-induced pulmonary fibrosis is unclear. METHODS: To evaluate the therapeutic efficacy of Trig in the context of silicosis-related pulmonary fibrosis, a mouse model of silicosis was utilized. The investigation seeks to elucidated Trig's impact on the progression of silica-induced pulmonary fibrosis by evaluating protein expression, mRNA levels and employing Hematoxylin and Eosin (H&E), Masson's trichrome, and Sirius Red staining. Subsequently, we explored the mechanism underlying of its functions. RESULTS: In vivo experiment, Trig has been demonstrated the significant efficacy in mitigating SiO2-induced silicosis and BLM-induced pulmonary fibrosis, as evidenced by improved histochemical staining and reduced fibrotic marker expressions. Additionally, we showed that the differentiation of fibroblast to myofibroblast was imped in Trig + SiO2 group. In terms of mechanism, we obtained in vitro evidence that Trig inhibited fibroblast-to-myofibroblast differentiation by repressing TGF-ß/Smad signaling according to the in vitro evidence. Notably, our finding indicated that Trig seemed to be safe in mice and fibroblasts. CONCLUSION: In summary, Trig attenuated the severity of silicosis-related pulmonary fibrosis by alleviating the differentiation of myofibroblasts, indicating the development of novel therapeutic approaches for silicosis fibrosis.


Subject(s)
Alkaloids , Cell Differentiation , Fibroblasts , Mice, Inbred C57BL , Myofibroblasts , Pulmonary Fibrosis , Silicon Dioxide , Silicosis , Animals , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/prevention & control , Alkaloids/pharmacology , Silicon Dioxide/toxicity , Mice , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Myofibroblasts/pathology , Cell Differentiation/drug effects , Silicosis/pathology , Silicosis/metabolism , Silicosis/drug therapy , Male
14.
Gene ; 927: 148730, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-38944165

ABSTRACT

Hypertrophic scar (HS) presents a significant clinical challenge, frequently arising as a fibrotic sequela of burn injuries and trauma. Characterized by the aberrant activation and proliferation of myofibroblasts, HS lacks a targeted therapeutic approach to effectively reduce this dysregulation. This study offers novel evidence of upregulated expression of CD248 in HS tissues compared to normal skin (NS) tissues. Specifically, the expression of CD248 was predominantly localized to α-SMA+-myofibroblasts in the dermis. To explain the functional role of CD248 in dermal myofibroblast activity, we employed a targeted anti-CD248 antibody, IgG78. Both CD248 intervention and IgG78 treatment effectively suppressed the proliferative, migratory, and ECM-synthesizing activities of myofibroblasts isolated from HS dermis. In addition, IgG78 administration significantly attenuated HS formation in an in vivo rabbit ear model. The LC/MS analysis coupled with co-immunoprecipitation of HS tissues indicated a direct interaction between CD248 and the ECM components Fibronectin (FN) and Collagen I (COL I). These findings collectively suggest that CD248 may function as a pro-fibrotic factor in HS development through its interaction with ECM constituents. The utilization of an anti-CD248 antibody, such as IgG78, represents a promising novel therapeutic strategy for the treatment of HS.


Subject(s)
Antigens, CD , Cicatrix, Hypertrophic , Extracellular Matrix , Fibronectins , Myofibroblasts , Animals , Cicatrix, Hypertrophic/metabolism , Cicatrix, Hypertrophic/pathology , Rabbits , Myofibroblasts/metabolism , Myofibroblasts/pathology , Humans , Extracellular Matrix/metabolism , Antigens, CD/metabolism , Antigens, CD/genetics , Fibronectins/metabolism , Cell Proliferation , Male , Collagen Type I/metabolism , Collagen Type I/genetics , Female , Cell Movement , Adult , Cells, Cultured , Actins/metabolism
15.
Life Sci ; 351: 122805, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38851422

ABSTRACT

AIMS: Heart failure (HF) is one of the most devastating consequences of cardiovascular diseases. Regardless of etiology, cardiac fibrosis is present and promotes the loss of heart function in HF patients. Cardiac resident fibroblasts, in response to a host of pro-fibrogenic stimuli, trans-differentiate into myofibroblasts to mediate cardiac fibrosis, the underlying mechanism of which remains incompletely understood. METHODS: Fibroblast-myofibroblast transition was induced in vitro by exposure to transforming growth factor (TGF-ß). Cardiac fibrosis was induced in mice by either transverse aortic constriction (TAC) or by chronic infusion with angiotensin II (Ang II). RESULTS: Through bioinformatic screening, we identified Kruppel-like factor 6 (KLF6) as a transcription factor preferentially up-regulated in cardiac fibroblasts from individuals with non-ischemic cardiomyopathy (NICM) compared to the healthy donors. Further analysis showed that nuclear factor kappa B (NF-κB) bound to the KLF6 promoter and mediated KLF6 trans-activation by pro-fibrogenic stimuli. KLF6 knockdown attenuated whereas KLF6 over-expression enhanced TGF-ß induced fibroblast-myofibroblast transition in vitro. More importantly, myofibroblast-specific KLF6 depletion ameliorated cardiac fibrosis and rescued heart function in mice subjected to the TAC procedure or chronic Ang II infusion. SIGNIFICANCE: In conclusion, our data support a role for KLF6 in cardiac fibrosis.


Subject(s)
Fibroblasts , Fibrosis , Kruppel-Like Factor 6 , Mice, Inbred C57BL , Myofibroblasts , Animals , Kruppel-Like Factor 6/metabolism , Kruppel-Like Factor 6/genetics , Fibrosis/metabolism , Mice , Humans , Male , Fibroblasts/metabolism , Myofibroblasts/metabolism , Myofibroblasts/pathology , Angiotensin II/pharmacology , Myocardium/metabolism , Myocardium/pathology , Transforming Growth Factor beta/metabolism , NF-kappa B/metabolism , Cells, Cultured , Kruppel-Like Transcription Factors/metabolism , Kruppel-Like Transcription Factors/genetics , Heart Failure/metabolism , Heart Failure/pathology , Heart Failure/genetics
16.
Circ Res ; 135(3): 453-469, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-38899461

ABSTRACT

BACKGROUND: Cardiac fibroblast activation contributes to adverse remodeling, fibrosis, and dysfunction in the pressure-overloaded heart. Although early fibroblast TGF-ß (transforming growth factor-ß)/Smad (small mother against decapentaplegic)-3 activation protects the pressure-overloaded heart by preserving the matrix, sustained TGF-ß activation is deleterious, accentuating fibrosis and dysfunction. Thus, endogenous mechanisms that negatively regulate the TGF-ß response in fibroblasts may be required to protect from progressive fibrosis and adverse remodeling. We hypothesized that Smad7, an inhibitory Smad that restrains TGF-ß signaling, may be induced in the pressure-overloaded myocardium and may regulate fibrosis, remodeling, and dysfunction. METHODS: The effects of myofibroblast-specific Smad7 loss were studied in a mouse model of transverse aortic constriction, using echocardiography, histological analysis, and molecular analysis. Proteomic studies in S7KO (Smad7 knockout) and overexpressing cells were used to identify fibroblast-derived mediators modulated by Smad7. In vitro experiments using cultured cardiac fibroblasts, fibroblasts populating collagen lattices, and isolated macrophages were used to dissect the molecular signals responsible for the effects of Smad7. RESULTS: Following pressure overload, Smad7 was upregulated in cardiac myofibroblasts. TGF-ß and angiotensin II stimulated fibroblast Smad7 upregulation via Smad3, whereas GDF15 (growth differentiation factor 15) induced Smad7 through GFRAL (glial cell line-derived neurotrophic factor family receptor α-like). MFS7KO (myofibroblast-specific S7KO) mice had increased mortality, accentuated systolic dysfunction and dilative remodeling, and accelerated diastolic dysfunction in response to transverse aortic constriction. Increased dysfunction in MFS7KO hearts was associated with accentuated fibrosis and increased MMP (matrix metalloproteinase)-2 activity and collagen denaturation. Secretomic analysis showed that Smad7 loss accentuates secretion of structural collagens and matricellular proteins and markedly increases MMP2 secretion. In contrast, Smad7 overexpression reduced MMP2 levels. In fibroblasts populating collagen lattices, the effects of Smad7 on fibroblast-induced collagen denaturation and pad contraction were partly mediated via MMP2 downregulation. Surprisingly, MFS7KO mice also exhibited significant macrophage expansion caused by paracrine actions of Smad7 null fibroblasts that stimulate macrophage proliferation and fibrogenic activation. Macrophage activation involved the combined effects of the fibroblast-derived matricellular proteins CD5L (CD5 antigen-like), SPARC (secreted protein acidic and rich in cysteine), CTGF (connective tissue growth factor), ECM1 (extracellular matrix protein 1), and TGFBI (TGFB induced). CONCLUSIONS: The antifibrotic effects of Smad7 in the pressure-overloaded heart protect from dysfunction and involve not only reduction in collagen deposition but also suppression of MMP2-mediated matrix denaturation and paracrine effects that suppress macrophage activation through inhibition of matricellular proteins.


Subject(s)
Fibrosis , Mice, Knockout , Myofibroblasts , Smad7 Protein , Ventricular Remodeling , Animals , Smad7 Protein/metabolism , Smad7 Protein/genetics , Mice , Myofibroblasts/metabolism , Myofibroblasts/pathology , Cells, Cultured , Mice, Inbred C57BL , Transforming Growth Factor beta/metabolism , Male , Fibroblasts/metabolism , Fibroblasts/pathology , Signal Transduction , Myocardium/metabolism , Myocardium/pathology
17.
Sci Adv ; 10(25): eadk8501, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38905342

ABSTRACT

Single-cell technology has allowed researchers to probe tissue complexity and dynamics at unprecedented depth in health and disease. However, the generation of high-dimensionality single-cell atlases and virtual three-dimensional tissues requires integrated reference maps that harmonize disparate experimental designs, analytical pipelines, and taxonomies. Here, we present a comprehensive single-cell transcriptome integration map of cardiac fibrosis, which underpins pathophysiology in most cardiovascular diseases. Our findings reveal similarity between cardiac fibroblast (CF) identities and dynamics in ischemic versus pressure overload models of cardiomyopathy. We also describe timelines for commitment of activated CFs to proliferation and myofibrogenesis, profibrotic and antifibrotic polarization of myofibroblasts and matrifibrocytes, and CF conservation across mouse and human healthy and diseased hearts. These insights have the potential to inform knowledge-based therapies.


Subject(s)
Fibroblasts , Fibrosis , Single-Cell Analysis , Transcriptome , Animals , Single-Cell Analysis/methods , Humans , Fibroblasts/metabolism , Mice , Myocardium/metabolism , Myocardium/pathology , Myofibroblasts/metabolism , Myofibroblasts/pathology , Gene Expression Profiling
18.
J Clin Invest ; 134(10)2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38747285

ABSTRACT

Transforming growth factor ß (TGF-ß) signaling is a core pathway of fibrosis, but the molecular regulation of the activation of latent TGF-ß remains incompletely understood. Here, we demonstrate a crucial role of WNT5A/JNK/ROCK signaling that rapidly coordinates the activation of latent TGF-ß in fibrotic diseases. WNT5A was identified as a predominant noncanonical WNT ligand in fibrotic diseases such as systemic sclerosis, sclerodermatous chronic graft-versus-host disease, and idiopathic pulmonary fibrosis, stimulating fibroblast-to-myofibroblast transition and tissue fibrosis by activation of latent TGF-ß. The activation of latent TGF-ß requires rapid JNK- and ROCK-dependent cytoskeletal rearrangements and integrin αV (ITGAV). Conditional ablation of WNT5A or its downstream targets prevented activation of latent TGF-ß, rebalanced TGF-ß signaling, and ameliorated experimental fibrosis. We thus uncovered what we believe to be a novel mechanism for the aberrant activation of latent TGF-ß in fibrotic diseases and provided evidence for targeting WNT5A/JNK/ROCK signaling in fibrotic diseases as a new therapeutic approach.


Subject(s)
Fibroblasts , Fibrosis , Transforming Growth Factor beta , Wnt-5a Protein , rho-Associated Kinases , Wnt-5a Protein/metabolism , Wnt-5a Protein/genetics , Animals , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/genetics , Mice , Humans , Fibroblasts/metabolism , Fibroblasts/pathology , rho-Associated Kinases/metabolism , rho-Associated Kinases/genetics , Scleroderma, Systemic/pathology , Scleroderma, Systemic/metabolism , Scleroderma, Systemic/genetics , Mice, Knockout , Wnt Proteins/metabolism , Wnt Proteins/genetics , MAP Kinase Signaling System , Myofibroblasts/metabolism , Myofibroblasts/pathology , Signal Transduction , Idiopathic Pulmonary Fibrosis/pathology , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/genetics
19.
Am J Surg Pathol ; 48(8): 1005-1016, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38717131

ABSTRACT

The number of recognized sarcoma types harboring targetable molecular alterations continues to increase. Here we present 25 examples of a distinctive myofibroblastic tumor, provisionally termed "myxoid inflammatory myofibroblastic sarcoma," which might be related to inflammatory myofibroblastic tumor, and which occurred in 13 males (52%) and 12 females at a median age of 37 years (range: 7 to 79 years). Primary tumor sites were peritoneum (18 patients; 72%), paratesticular (2; 8%), chest wall (1), upper extremity (1), esophagus (1), retroperitoneum (1), and uterus (1). Nine peritoneal tumors (50%) were multifocal at presentation; all other tumors were unifocal. Tumors showed bland-to-mildly-atypical neoplastic myofibroblasts in a myxoid stroma, with prominent inflammatory infiltrates in 22 cases (88%). Most tumors showed delicate branching stromal vessels like those of myxoid liposarcoma, and most showed infiltrative growth through non-neoplastic tissue. Immunohistochemistry demonstrated expression of SMA (19/25 tumors; 76%), desmin (13/22; 59%), and CD30 (5/11; 45%), while ALK was expressed in 1 tumor (of 25; 4%) that was negative for ALK rearrangement. Sequencing of 11 tumors showed seven to harbor tyrosine kinase fusions (4 PDGFRB , 2 PML :: JAK1 , 1 SEC31A :: PDGFRA ). Two instead harbored hot spot KRAS mutations (G12V and Q61H), and 2 were negative for known driving alterations. Clinical follow-up was available for 18 patients (72%; median: 2.7 years; range: 4 mo-12.3 years). Nine patients (50%) were alive with no evidence of disease, 5 (28%) died of disease, and 4 (22%) were alive with disease. Seven patients (39%) experienced peritoneal relapse or distant metastasis. Two patients showed disease progression on conventional, nontargeted chemotherapy. The patient whose tumor harbored SEC31A :: PDGFRA was treated after multiple relapses with imatinib and sunitinib therapy, with progression-free periods of 5 and 2 years, respectively. Despite its bland appearance, myxoid inflammatory myofibroblastic sarcoma harbors a significant risk for disseminated disease, particularly when it occurs in the peritoneum. Targeted therapy could be considered for patients with disseminated disease.


Subject(s)
Biomarkers, Tumor , Myofibroblasts , Humans , Male , Female , Adult , Middle Aged , Aged , Adolescent , Young Adult , Child , Biomarkers, Tumor/genetics , Biomarkers, Tumor/analysis , Myofibroblasts/pathology , Myofibroblasts/chemistry , Soft Tissue Neoplasms/pathology , Soft Tissue Neoplasms/genetics , Soft Tissue Neoplasms/mortality , Soft Tissue Neoplasms/therapy , Sarcoma/pathology , Sarcoma/genetics , Sarcoma/chemistry , Sarcoma/mortality , Immunohistochemistry
20.
Commun Biol ; 7(1): 544, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38714800

ABSTRACT

Numerous myofibroblasts are arisen from endothelial cells (ECs) through endothelial to mesenchymal transition (EndMT) triggered by TGF-ß. However, the mechanism of ECs transforms to a different subtype, or whether there exists an intermediate state of ECs remains unclear. In present study, we demonstrate Midkine (MDK) mainly expressed by CD31 + ACTA2+ECs going through partial EndMT contribute greatly to myofibroblasts by spatial and single-cell transcriptomics. MDK is induced in TGF-ß treated ECs, which upregulates C/EBPß and increases EndMT genes, and these effects could be reversed by siMDK. Mechanistically, MDK promotes the binding ability of C/EBPß with ACTA2 promoter by stabilizing the C/EBPß protein. In vivo, knockout of Mdk or conditional knockout of Mdk in ECs reduces EndMT markers and significantly reverses fibrogenesis. In conclusion, our study provides a mechanistic link between the induction of EndMT by TGF-ß and MDK, which suggests that blocking MDK provides potential therapeutic strategies for renal fibrosis.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta , Endothelial-Mesenchymal Transition , Fibrosis , Midkine , Animals , Humans , Male , Mice , CCAAT-Enhancer-Binding Protein-beta/metabolism , CCAAT-Enhancer-Binding Protein-beta/genetics , Endothelial Cells/metabolism , Endothelial Cells/pathology , Kidney/metabolism , Kidney/pathology , Kidney Diseases/metabolism , Kidney Diseases/pathology , Kidney Diseases/genetics , Mice, Inbred C57BL , Mice, Knockout , Midkine/metabolism , Midkine/genetics , Myofibroblasts/metabolism , Myofibroblasts/pathology , Transforming Growth Factor beta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL