1
|
Yang H, Xiang Y, Wang J, Ke Z, Zhou W, Yin X, Zhang M, Chen Z. Modulating the blood-brain barrier in CNS disorders: A review of the therapeutic implications of secreted protein acidic and rich in cysteine (SPARC). Int J Biol Macromol 2025; 288:138747. [PMID: 39674451 DOI: 10.1016/j.ijbiomac.2024.138747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 12/09/2024] [Accepted: 12/11/2024] [Indexed: 12/16/2024]
Abstract
Secreted protein acidic and rich in cysteine (SPARC), an essential stromal cell protein, plays a crucial role in angiogenesis and maintaining endothelial barrier function. This protein is expressed by diverse cell types, including endothelial cells, fibroblasts, and macrophages, with increased expression found in regions of tissues undergoing active remodeling, repair, and proliferation. The role of SPARC in non-neural tissues is of significant interest. In the central nervous system (CNS), SPARC is highly expressed in blood vessels during early development. It becomes down-regulated as the brain matures, a pattern consistent with its role in angiogenesis and blood-brain barrier (BBB) establishment. In this review, we explore the multifaceted roles of SPARC in regulating CNS disorders, particularly its action in angiogenesis, inflammatory responses, neural system development and repair, barrier establishment, maintenance of BBB function, and the pathogenesis of CNS disorders triggered by BBB dysfunction.
Collapse
Affiliation(s)
- Hui Yang
- Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi 332000, China; Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China; School of Basic Medicine, Jiujiang University, Jiujiang, Jiangxi 332000, China
| | - Yuanyuan Xiang
- Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi 332000, China
| | - Jiaxuan Wang
- Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi 332000, China; Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China; School of Basic Medicine, Jiujiang University, Jiujiang, Jiangxi 332000, China
| | - Zunliang Ke
- Department of Neurosurgery, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi 332000, China
| | - Weixin Zhou
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China
| | - Xiaoping Yin
- Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi 332000, China; Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China
| | - Manqing Zhang
- School of Basic Medicine, Jiujiang University, Jiujiang, Jiangxi 332000, China.
| | - Zhiying Chen
- Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi 332000, China; Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China.
| |
Collapse
|
2
|
Lee B, Jo D, Park J, Kim OY, Song J. Gut microbiota and their relationship with circulating adipokines in an acute hepatic encephalopathy mouse model induced by surgical bile duct ligation. Heliyon 2024; 10:e38534. [PMID: 39391493 PMCID: PMC11466606 DOI: 10.1016/j.heliyon.2024.e38534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/11/2024] [Accepted: 09/25/2024] [Indexed: 10/12/2024] Open
Abstract
Background and aims Various studies have shown the importance of the gut microbiota in human health. However, little is known about gut microbiome patterns and their effect on circulating adipo-myokine levels in hepatic encephalopathy (HE). We investigated the relationship between the gut microbiota and adipo-myokine levels using a mouse model of HE induced by surgical bile duct ligation (BDL). Methods and results Wild-type C57BL/6J mice were subjected to sham surgery or BDL. Severe body weight loss, suppressed feed intake, and liver failure were observed in BDL mice compared with sham control mice. Additionally, changes in gut microbial communities and serum adipo-myokine levels were noted in BDL mice. In the BDL mouse gut, we identified 15 differentially abundant taxa including the phylum Verrucomicrobiota, the classes Actinomycetes and Verrucomicrobiae, the order Verrucomicrobiales, the families Akkermansiaceae, Bacteroidaceae, Rikenellaceae, and Oscillospiraceae, the genera Alistipes, Akkermansia, Muribaculum, and Phocaeicola, and the species Akkermansia muciniphila, Alistipes okayasuensis, and Muribaculum gordoncarteri by LEfSe analysis (LDA score≥4.0). Higher levels of certain adipo-myokines such as BDNF were detected in the serum of BDL mice. Spearman correlation analysis revealed that certain adipo-myokines (e.g., FSTL1) were positively correlated with the class Actinomycetes, the family Rikenellaceae, the genus Alistipes, and the species Alistipes okayasuensis. Interestingly, A. okayasuensis and M. gordoncarteri, recently isolated microbes, showed richness in the gut of BDL mice and demonstrated positive correlations with adipo-myokines such as FGF21. Conclusions Overall, our results suggest that alteration of the gut microbiota in patients with HE may be closely correlated to the levels of adipo-myokines in the blood.
Collapse
Affiliation(s)
- Bokyung Lee
- Department of Food Science and Nutrition, Dong A University, Sahagu, Nakdongdaero 550 beon-gil, 49315, Busan, Republic of Korea
| | - Danbi Jo
- Department of Anatomy, Chonnam National University Medical School, Hwasun, 58128, Jeollanam-do, Republic of Korea
| | - Jihyun Park
- Department of Health Sciences, Graduate School of Dong-A University, Sahagu, Nakdongdaero 550 beon-gil, 49315, Busan, Republic of Korea
| | - Oh Yoen Kim
- Department of Food Science and Nutrition, Dong A University, Sahagu, Nakdongdaero 550 beon-gil, 49315, Busan, Republic of Korea
- Department of Health Sciences, Graduate School of Dong-A University, Sahagu, Nakdongdaero 550 beon-gil, 49315, Busan, Republic of Korea
| | - Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Hwasun, 58128, Jeollanam-do, Republic of Korea
| |
Collapse
|
3
|
Ruiz-Blázquez P, Fernández-Fernández M, Pistorio V, Martinez-Sanchez C, Costanzo M, Iruzubieta P, Zhuravleva E, Cacho-Pujol J, Ariño S, Del Castillo-Cruz A, Núñez S, Andersen JB, Ruoppolo M, Crespo J, García-Ruiz C, Pavone LM, Reinheckel T, Sancho-Bru P, Coll M, Fernández-Checa JC, Moles A. Cathepsin D is essential for the degradomic shift of macrophages required to resolve liver fibrosis. Mol Metab 2024; 87:101989. [PMID: 39019115 PMCID: PMC11327474 DOI: 10.1016/j.molmet.2024.101989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/03/2024] [Accepted: 07/08/2024] [Indexed: 07/19/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Fibrosis contributes to 45% of deaths in industrialized nations and is characterized by an abnormal accumulation of extracellular matrix (ECM). There are no specific anti-fibrotic treatments for liver fibrosis, and previous unsuccessful attempts at drug development have focused on preventing ECM deposition. Because liver fibrosis is largely acknowledged to be reversible, regulating fibrosis resolution could offer novel therapeutical options. However, little is known about the mechanisms controlling ECM remodeling during resolution. Changes in proteolytic activity are essential for ECM homeostasis and macrophages are an important source of proteases. Herein, in this study we evaluate the role of macrophage-derived cathepsin D (CtsD) during liver fibrosis. METHODS CtsD expression and associated pathways were characterized in single-cell RNA sequencing and transcriptomic datasets in human cirrhosis. Liver fibrosis progression, reversion and functional characterization were assessed in novel myeloid-CtsD and hepatocyte-CtsD knock-out mice. RESULTS Analysis of single-cell RNA sequencing datasets demonstrated CtsD was expressed in macrophages and hepatocytes in human cirrhosis. Liver fibrosis progression, reversion and functional characterization were assessed in novel myeloid-CtsD (CtsDΔMyel) and hepatocyte-CtsD knock-out mice. CtsD deletion in macrophages, but not in hepatocytes, resulted in enhanced liver fibrosis. Both inflammatory and matrisome proteomic signatures were enriched in fibrotic CtsDΔMyel livers. Besides, CtsDΔMyel liver macrophages displayed functional, phenotypical and secretomic changes, which resulted in a degradomic phenotypical shift, responsible for the defective proteolytic processing of collagen I in vitro and impaired collagen remodeling during fibrosis resolution in vivo. Finally, CtsD-expressing mononuclear phagocytes of cirrhotic human livers were enriched in lysosomal and ECM degradative signaling pathways. CONCLUSIONS Our work describes for the first-time CtsD-driven lysosomal activity as a central hub for restorative macrophage function during fibrosis resolution and opens new avenues to explore their degradome landscape to inform drug development.
Collapse
Affiliation(s)
- Paloma Ruiz-Blázquez
- Institute of Biomedical Research of Barcelona, Spanish National Research Council, Barcelona, Spain; CiberEHD, Spain; University of Barcelona, Barcelona, Spain; IDIBAPS, Barcelona, Spain
| | - María Fernández-Fernández
- Institute of Biomedical Research of Barcelona, Spanish National Research Council, Barcelona, Spain; CiberEHD, Spain; University of Barcelona, Barcelona, Spain; IDIBAPS, Barcelona, Spain
| | - Valeria Pistorio
- Institute of Biomedical Research of Barcelona, Spanish National Research Council, Barcelona, Spain; Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy; Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine (CRSA), Paris, France
| | | | - Michele Costanzo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy; CEINGE-Biotecnologie Avanzate Franco Salvatore s.c.ar.l., Naples, Italy
| | - Paula Iruzubieta
- Department of Gastroenterology and Hepatology, Marqués de Valdecilla University Hospital, Research Institute Marqués de Valdecilla (IDIVAL), Santander, Spain
| | - Ekaterina Zhuravleva
- Biotech Research and Innovation Centre (BRIC), Department of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; LEO Foundation Skin Immunology Research Center (SIC), Department of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Júlia Cacho-Pujol
- Institute of Biomedical Research of Barcelona, Spanish National Research Council, Barcelona, Spain; University of Barcelona, Barcelona, Spain; IDIBAPS, Barcelona, Spain
| | - Silvia Ariño
- CiberEHD, Spain; University of Barcelona, Barcelona, Spain; IDIBAPS, Barcelona, Spain
| | | | | | - Jesper B Andersen
- Biotech Research and Innovation Centre (BRIC), Department of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Margherita Ruoppolo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy; CEINGE-Biotecnologie Avanzate Franco Salvatore s.c.ar.l., Naples, Italy
| | - Javier Crespo
- Department of Gastroenterology and Hepatology, Marqués de Valdecilla University Hospital, Research Institute Marqués de Valdecilla (IDIVAL), Santander, Spain
| | - Carmen García-Ruiz
- Institute of Biomedical Research of Barcelona, Spanish National Research Council, Barcelona, Spain; CiberEHD, Spain; IDIBAPS, Barcelona, Spain; USC Research Center for ALPD, Los Angeles, United States; Associated Unit IIBB-IMIM, Barcelona, Spain
| | - Luigi Michele Pavone
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Thomas Reinheckel
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, Albert-Ludwigs-University, Freiburg, Germany; German Cancer Consortium (DKTK), DKFZ Partner Site Freiburg, Germany; Center for Biological Signaling Studies BIOSS, University of Freiburg, Germany
| | - Pau Sancho-Bru
- CiberEHD, Spain; University of Barcelona, Barcelona, Spain; IDIBAPS, Barcelona, Spain
| | - Mar Coll
- CiberEHD, Spain; University of Barcelona, Barcelona, Spain; IDIBAPS, Barcelona, Spain; Medicine Department, Faculty of Medicine, University of Barcelona, Spain
| | - José C Fernández-Checa
- Institute of Biomedical Research of Barcelona, Spanish National Research Council, Barcelona, Spain; CiberEHD, Spain; IDIBAPS, Barcelona, Spain; USC Research Center for ALPD, Los Angeles, United States; Associated Unit IIBB-IMIM, Barcelona, Spain
| | - Anna Moles
- Institute of Biomedical Research of Barcelona, Spanish National Research Council, Barcelona, Spain; CiberEHD, Spain; IDIBAPS, Barcelona, Spain; Associated Unit IIBB-IMIM, Barcelona, Spain.
| |
Collapse
|
4
|
Coler-Reilly A, Pincus Z, Scheller EL, Civitelli R. Six drivers of aging identified among genes differentially expressed with age. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.02.606402. [PMID: 39149379 PMCID: PMC11326176 DOI: 10.1101/2024.08.02.606402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Many studies have compared gene expression in young and old samples to gain insights on aging, the primary risk factor for most major chronic diseases. However, these studies only describe associations, failing to distinguish drivers of aging from compensatory geroprotective responses and incidental downstream effects. Here, we introduce a workflow to characterize the causal effects of differentially expressed genes on lifespan. First, we performed a meta-analysis of 25 gene expression datasets comprising samples of various tissues from healthy, untreated adult mammals (humans, dogs, and rodents) at two distinct ages. We ranked each gene according to the number of distinct datasets in which the gene was differentially expressed with age in a consistent direction. The top age-upregulated genes were TMEM176A, EFEMP1, CP, and HLA-A; the top age-downregulated genes were CA4, SIAH, SPARC, and UQCR10. Second, the effects of the top ranked genes on lifespan were measured by applying post-developmental RNA interference of the corresponding ortholog in the nematode C. elegans (two trials, with roughly 100 animals per genotype per trial). Out of 10 age-upregulated and 9 age-downregulated genes that were tested, two age-upregulated genes (csp-3/CASP1 and spch-2/RSRC1) and four age-downregulated genes (C42C1.8/DIRC2, ost-1/SPARC, fzy-1/CDC20, and cah-3/CA4) produced significant and reproducible lifespan extension. Notably, the data do not suggest that the direction of differential expression with age is predictive of the effect on lifespan. Our study provides novel insight into the relationship between differential gene expression and aging phenotypes, pilots an unbiased workflow that can be easily repeated and expanded, and pinpoints six genes with evolutionarily conserved, causal roles in the aging process for further study.
Collapse
Affiliation(s)
- Ariella Coler-Reilly
- Division of Bone and Mineral Diseases, Musculoskeletal Research Center
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Medical Scientist Training Program, Washington University School of Medicine, St. Louis, MO, USA
| | - Zachary Pincus
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Erica L. Scheller
- Division of Bone and Mineral Diseases, Musculoskeletal Research Center
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Cell Biology and Physiology; Washington University School of Medicine, St. Louis, MO, USA
| | - Roberto Civitelli
- Division of Bone and Mineral Diseases, Musculoskeletal Research Center
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Cell Biology and Physiology; Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
5
|
Atorrasagasti C, Onorato AM, Mazzolini G. The role of SPARC (secreted protein acidic and rich in cysteine) in the pathogenesis of obesity, type 2 diabetes, and non-alcoholic fatty liver disease. J Physiol Biochem 2023; 79:815-831. [PMID: 36018492 DOI: 10.1007/s13105-022-00913-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 07/18/2022] [Indexed: 11/28/2022]
Abstract
Secreted protein acidic and rich in cysteine (SPARC) is an extracellular matrix glycoprotein with pleiotropic functions, which is expressed in adipose, hepatic, muscular, and pancreatic tissue. Particularly, several studies demonstrated that SPARC is an important player in the context of obesity, diabetes, and fatty liver disease including advanced hepatic fibrosis and hepatocellular carcinoma. Evidence in murine and human samples indicates that SPARC is involved in adipogenesis, cellular metabolism, extracellular matrix modulation, glucose and lipid metabolism, among others. Furthermore, studies in SPARC knockout mouse model showed that SPARC contributes to adipose tissue formation, non-alcoholic fatty liver disease (NAFLD), and diabetes. Hence, SPARC may represent a novel and interesting target protein for future therapeutic interventions or a biomarker of disease progression. This review summarizes the role of SPARC in the pathophysiology of obesity, and extensively revised SPARC functions in physiological and pathological adipose tissue deposition, muscle metabolism, liver, and diabetes-related pathways.
Collapse
Affiliation(s)
- Catalina Atorrasagasti
- Gene Therapy Laboratory, Facultad de Ciencias Biomédicas, Instituto de Investigaciones en Medicina Traslacional, CONICET- Universidad Austral, Av. Pte. Perón 1500 (B1629AHJ) Derqui-Pilar, Buenos Aires, Argentina.
| | - Agostina M Onorato
- Gene Therapy Laboratory, Facultad de Ciencias Biomédicas, Instituto de Investigaciones en Medicina Traslacional, CONICET- Universidad Austral, Av. Pte. Perón 1500 (B1629AHJ) Derqui-Pilar, Buenos Aires, Argentina
| | - Guillermo Mazzolini
- Gene Therapy Laboratory, Facultad de Ciencias Biomédicas, Instituto de Investigaciones en Medicina Traslacional, CONICET- Universidad Austral, Av. Pte. Perón 1500 (B1629AHJ) Derqui-Pilar, Buenos Aires, Argentina.
- Liver Unit, Hospital Universitario Austral, Universidad Austral, Av. Pte. Perón 1500 (B1629AHJ) Derqui-Pilar, Buenos Aires, Argentina.
| |
Collapse
|
6
|
Onorato AM, Lameroli Mauriz L, Bayo J, Fiore E, Cantero MJ, Bueloni B, García M, Lagües C, Martínez-Duartez P, Menaldi G, Paleari N, Atorrasagasti C, Mazzolini GD. Hepatic SPARC Expression Is Associated with Inflammasome Activation during the Progression of Non-Alcoholic Fatty Liver Disease in Both Mice and Morbidly Obese Patients. Int J Mol Sci 2023; 24:14843. [PMID: 37834291 PMCID: PMC10573696 DOI: 10.3390/ijms241914843] [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: 09/02/2023] [Revised: 09/23/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023] Open
Abstract
The severity of non-alcoholic fatty liver disease (NAFLD) ranges from simple steatosis to steatohepatitis, and it is not yet clearly understood which patients will progress to liver fibrosis or cirrhosis. SPARC (Secreted Protein Acidic and Rich in Cysteine) has been involved in NAFLD pathogenesis in mice and humans. The aim of this study was to investigate the role of SPARC in inflammasome activation, and to evaluate the relationship between the hepatic expression of inflammasome genes and the biochemical and histological characteristics of NAFLD in obese patients. In vitro studies were conducted in a macrophage cell line and primary hepatocyte cultures to assess the effect of SPARC on inflammasome. A NAFLD model was established in SPARC knockout (SPARC-/-) and SPARC+/+ mice to explore inflammasome activation. A hepatic RNAseq database from NAFLD patients was analyzed to identify genes associated with SPARC expression. The results were validated in a prospective cohort of 59 morbidly obese patients with NAFLD undergoing bariatric surgery. Our results reveal that SPARC alone or in combination with saturated fatty acids promoted IL-1β expression in cell cultures. SPARC-/- mice had reduced hepatic inflammasome activation during the progression of NAFLD. NAFLD patients showed increased expression of SPARC, NLRP3, CASP1, and IL-1β. Gene ontology analysis revealed that genes positively correlated with SPARC are linked to inflammasome-related pathways during the progression of the disease, enabling the differentiation of patients between steatosis and steatohepatitis. In conclusion, SPARC may play a role in hepatic inflammasome activation in NAFLD.
Collapse
Affiliation(s)
- Agostina M. Onorato
- Gene Therapy Laboratory, Instituto de Investigaciones en Medicina Traslacional, Facultad de Ciencias Biomédicas, CONICET-Universidad Austral, Av. Pte. Perón 1500, Pilar B1629AHJ, Argentina; (A.M.O.); (L.L.M.); (J.B.); (E.F.); (M.J.C.); (B.B.); (M.G.)
| | - Lucía Lameroli Mauriz
- Gene Therapy Laboratory, Instituto de Investigaciones en Medicina Traslacional, Facultad de Ciencias Biomédicas, CONICET-Universidad Austral, Av. Pte. Perón 1500, Pilar B1629AHJ, Argentina; (A.M.O.); (L.L.M.); (J.B.); (E.F.); (M.J.C.); (B.B.); (M.G.)
| | - Juan Bayo
- Gene Therapy Laboratory, Instituto de Investigaciones en Medicina Traslacional, Facultad de Ciencias Biomédicas, CONICET-Universidad Austral, Av. Pte. Perón 1500, Pilar B1629AHJ, Argentina; (A.M.O.); (L.L.M.); (J.B.); (E.F.); (M.J.C.); (B.B.); (M.G.)
| | - Esteban Fiore
- Gene Therapy Laboratory, Instituto de Investigaciones en Medicina Traslacional, Facultad de Ciencias Biomédicas, CONICET-Universidad Austral, Av. Pte. Perón 1500, Pilar B1629AHJ, Argentina; (A.M.O.); (L.L.M.); (J.B.); (E.F.); (M.J.C.); (B.B.); (M.G.)
| | - María José Cantero
- Gene Therapy Laboratory, Instituto de Investigaciones en Medicina Traslacional, Facultad de Ciencias Biomédicas, CONICET-Universidad Austral, Av. Pte. Perón 1500, Pilar B1629AHJ, Argentina; (A.M.O.); (L.L.M.); (J.B.); (E.F.); (M.J.C.); (B.B.); (M.G.)
| | - Barbara Bueloni
- Gene Therapy Laboratory, Instituto de Investigaciones en Medicina Traslacional, Facultad de Ciencias Biomédicas, CONICET-Universidad Austral, Av. Pte. Perón 1500, Pilar B1629AHJ, Argentina; (A.M.O.); (L.L.M.); (J.B.); (E.F.); (M.J.C.); (B.B.); (M.G.)
| | - Mariana García
- Gene Therapy Laboratory, Instituto de Investigaciones en Medicina Traslacional, Facultad de Ciencias Biomédicas, CONICET-Universidad Austral, Av. Pte. Perón 1500, Pilar B1629AHJ, Argentina; (A.M.O.); (L.L.M.); (J.B.); (E.F.); (M.J.C.); (B.B.); (M.G.)
| | - Cecilia Lagües
- Pathological Anatomy Department, Hospital Universitario Austral, Universidad Austral, Av. Pte. Perón 1500, Pilar B1629AHJ, Argentina
| | - Pedro Martínez-Duartez
- Bariatric and Metabolic Surgery Department, Hospital Universitario Austral, Universidad Austral, Av. Pte. Perón 1500, Pilar B1629AHJ, Argentina
| | - Gabriel Menaldi
- Bariatric and Metabolic Surgery Department, Hospital Universitario Austral, Universidad Austral, Av. Pte. Perón 1500, Pilar B1629AHJ, Argentina
| | - Nicolas Paleari
- Bariatric and Metabolic Surgery Department, Hospital Universitario Austral, Universidad Austral, Av. Pte. Perón 1500, Pilar B1629AHJ, Argentina
| | - Catalina Atorrasagasti
- Gene Therapy Laboratory, Instituto de Investigaciones en Medicina Traslacional, Facultad de Ciencias Biomédicas, CONICET-Universidad Austral, Av. Pte. Perón 1500, Pilar B1629AHJ, Argentina; (A.M.O.); (L.L.M.); (J.B.); (E.F.); (M.J.C.); (B.B.); (M.G.)
| | - Guillermo D. Mazzolini
- Gene Therapy Laboratory, Instituto de Investigaciones en Medicina Traslacional, Facultad de Ciencias Biomédicas, CONICET-Universidad Austral, Av. Pte. Perón 1500, Pilar B1629AHJ, Argentina; (A.M.O.); (L.L.M.); (J.B.); (E.F.); (M.J.C.); (B.B.); (M.G.)
- Liver Unit, Hospital Universitario Austral, Universidad Austral, Av. Pte. Perón 1500, Pilar B1629AHJ, Argentina
| |
Collapse
|
7
|
Tracking matricellular protein SPARC in extracellular vesicles as a non-destructive method to evaluate lipid-based antifibrotic treatments. Commun Biol 2022; 5:1155. [PMID: 36310239 PMCID: PMC9618575 DOI: 10.1038/s42003-022-04123-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 10/17/2022] [Indexed: 11/10/2022] Open
Abstract
Uncovering the complex cellular mechanisms underlying hepatic fibrogenesis could expedite the development of effective treatments and noninvasive diagnosis for liver fibrosis. The biochemical complexity of extracellular vesicles (EVs) and their role in intercellular communication make them an attractive tool to look for biomarkers as potential alternative to liver biopsies. We developed a solid set of methods to isolate and characterize EVs from differently treated human hepatic stellate cell (HSC) line LX-2, and we investigated their biological effect onto naïve LX-2, proving that EVs do play an active role in fibrogenesis. We mined our proteomic data for EV-associated proteins whose expression correlated with HSC treatment, choosing the matricellular protein SPARC as proof-of-concept for the feasibility of fluorescence nanoparticle-tracking analysis to determine an EV-based HSCs’ fibrogenic phenotype. We thus used EVs to directly evaluate the efficacy of treatment with S80, a polyenylphosphatidylcholines-rich lipid, finding that S80 reduces the relative presence of SPARC-positive EVs. Here we correlated the cellular response to lipid-based antifibrotic treatment to the relative presence of a candidate protein marker associated with the released EVs. Along with providing insights into polyenylphosphatidylcholines treatments, our findings pave the way for precise and less invasive diagnostic analyses of hepatic fibrogenesis. A method is developed to isolate and characterize extracellular vesicles (EVs) from human hepatic stellate cells and proteomics reveals that the matricellular protein SPARC may be used as an EV marker after lipid-based antifibrotic treatment.
Collapse
|
8
|
MacDonald WW, Swaminathan SS, Heo JY, Castillejos A, Hsueh J, Liu BJ, Jo D, Du A, Lee H, Kang MH, Rhee DJ. Effect of SPARC Suppression in Mice, Perfused Human Anterior Segments, and Trabecular Meshwork Cells. Invest Ophthalmol Vis Sci 2022; 63:8. [PMID: 35671048 PMCID: PMC9187959 DOI: 10.1167/iovs.63.6.8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Purpose Secreted protein, acidic and rich in cysteine (SPARC) elevates intraocular pressure (IOP), increases certain structural extracellular matrix (ECM) proteins in the juxtacanalicular trabecular meshwork (JCT), and decreases matrix metalloproteinase (MMP) protein levels in trabecular meshwork (TM) endothelial cells. We investigated SPARC as a potential target for lowering IOP. We hypothesized that suppressing SPARC will decrease IOP, decrease structural JCT ECM proteins, and alter the levels of MMPs and/or their inhibitors. Methods A lentivirus containing short hairpin RNA of human SPARC suppressed SPARC in mouse eyes and perfused cadaveric human anterior segments with subsequent IOP measurements. Immunohistochemistry determined structural correlates. Human TM cell cultures were treated with SPARC suppressing lentivirus. Quantitative reverse transcriptase polymerase chain reaction (PCR), immunoblotting, and zymography determined total RNA, relative protein levels, and MMP enzymatic activity, respectively. Results Suppressing SPARC decreased IOP in mouse eyes and perfused human anterior segments by approximately 20%. Histologically, this correlated to a decrease in collagen I, IV, and VI in both the mouse TM and human JCT regions; in the mouse, fibronectin was also decreased but not in the human. In TM cells, collagen I and IV, fibronectin, MMP-2, and tissue inhibitor of MMP-1 were decreased. Messenger RNA of the aforementioned genes was not changed. Plasminogen activator inhibitor 1 (PAI-1) was upregulated in vitro by quantitative PCR and immunoblotting. MMP-1 activity was reduced in vitro by zymography. Conclusions Suppressing SPARC decreased IOP in mice and perfused cadaveric human anterior segments corresponding to qualitative structural changes in the JCT ECM, which do not appear to be the result of transcription regulation.
Collapse
Affiliation(s)
- William W MacDonald
- Department of Ophthalmology & Visual Sciences, University Hospitals, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States
| | - Swarup S Swaminathan
- Department of Ophthalmology & Visual Sciences, University Hospitals, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States.,Department of Ophthalmology, Bascom Palmer Eye Institute, Miami, Florida, United States
| | - Jae Young Heo
- Department of Ophthalmology & Visual Sciences, University Hospitals, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States
| | - Alexandra Castillejos
- Department of Ophthalmology & Visual Sciences, University Hospitals, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States.,Department of Ophthalmology, Massachusetts Eye & Ear Infirmary, Boston, Massachusetts, United States
| | - Jessica Hsueh
- Department of Ophthalmology & Visual Sciences, University Hospitals, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States
| | - Brian J Liu
- Department of Ophthalmology & Visual Sciences, University Hospitals, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States
| | - Diane Jo
- Department of Ophthalmology & Visual Sciences, University Hospitals, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States
| | - Annie Du
- Department of Ophthalmology & Visual Sciences, University Hospitals, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States
| | - Hyunpil Lee
- Department of Ophthalmology & Visual Sciences, University Hospitals, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States
| | - Min Hyung Kang
- Department of Ophthalmology & Visual Sciences, University Hospitals, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States
| | - Douglas J Rhee
- Department of Ophthalmology & Visual Sciences, University Hospitals, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States
| |
Collapse
|
9
|
Stage-dependent expression of fibrogenic markers in alcohol-related liver disease. Pathol Res Pract 2022; 231:153798. [DOI: 10.1016/j.prp.2022.153798] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 02/02/2022] [Accepted: 02/04/2022] [Indexed: 02/07/2023]
|
10
|
Hu C, Yin L, Chen Z, Waldron RT, Lugea A, Lin Y, Zhai X, Wen L, Han YP, Pandol SJ, Deng L, Xia Q. The unique pancreatic stellate cell gene expression signatures are associated with the progression from acute to chronic pancreatitis. Comput Struct Biotechnol J 2021; 19:6375-6385. [PMID: 34938413 PMCID: PMC8649580 DOI: 10.1016/j.csbj.2021.11.031] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 11/16/2021] [Accepted: 11/20/2021] [Indexed: 02/05/2023] Open
Abstract
Early recognition of chronic pancreatitis (CP) is still lacking. In the setting of CP injury, activated pancreatic stellate cell (PSC) is the central mediator of pancreatic fibrosis. We systematically define highly and uniquely expressed PSC genes and show that these genes are enriched in pancreatic diseases. Unresolved or recurrent injury causes dysregulation of biological process following AP, which would cause CP. We demonstrated subset genes that may be associated with the progression from AP to CP. Furthermore, SPARC was identified as a candidate marker for the disease progression. Increased expression of SPARC and canonical PSC genes were verified during AP recovery, especially in recurrent AP mice models.
Chronic pancreatitis (CP) is characterized by irreversible fibro-inflammatory changes induced by pancreatic stellate cell (PSC). Unresolved or recurrent injury causes dysregulation of biological process following AP, which would cause CP. Here, we systematically identify genes whose expressions are unique to PSC by comparing transcriptome profiles among total pancreas, pancreatic stellate, acinar, islet and immune cells. We then identified candidate genes and correlated them with the pancreatic disease continuum by performing intersection analysis among total PSC and activated PSC genes, and genes persistently differentially expressed during acute pancreatitis (AP) recovery. Last, we examined the association between candidate genes and AP, and substantiated their potential as biomarkers in experimental AP and recurrent AP (RAP) models. A total of 68 genes were identified as highly and uniquely expressed in PSC. The PSC signatures were highly enriched with extracellular matrix remodeling genes and were significantly enriched in AP pancreas compared to healthy control tissues. Among PSC signature genes that comprised a fibrotic phenotype, 10 were persistently differentially expressed during AP recovery. SPARC was determined as a candidate marker for the pancreatic disease continuum, which was not only persistently differentially expressed even five days after AP injury, but also highly expressed in two clinical datasets of CP. Sparc was also validated as highly elevated in RAP compared to AP mice. This work highlights the unique transcriptional profiles of PSC. These PSC signatures’ expression may help to identify patients with high risk of AP progression to CP.
Collapse
Affiliation(s)
- Cheng Hu
- Department and Laboratory of Integrated Traditional Chinese and Western Medicine, Sichuan Provincial Pancreatitis Center and West China-Liverpool Biomedical Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Liyuan Yin
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Zhiyao Chen
- Department and Laboratory of Integrated Traditional Chinese and Western Medicine, Sichuan Provincial Pancreatitis Center and West China-Liverpool Biomedical Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Richard T Waldron
- Department of Medicine and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States.,Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Aurelia Lugea
- Department of Medicine and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States.,Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Yiyun Lin
- Graduate School of Biomedical Sciences, UT MD Anderson Cancer Center, University of Texas, Houston, TX, United States
| | - Xiaoqian Zhai
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Li Wen
- Department of Gastroenterology and Shanghai Key Laboratory of Pancreatic Disease, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuan-Ping Han
- Center for Growth, Metabolism and Aging, College of Life Sciences, Sichuan University, Chengdu, China
| | - Stephen J Pandol
- Department of Medicine and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States.,Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Lihui Deng
- Department and Laboratory of Integrated Traditional Chinese and Western Medicine, Sichuan Provincial Pancreatitis Center and West China-Liverpool Biomedical Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Qing Xia
- Department and Laboratory of Integrated Traditional Chinese and Western Medicine, Sichuan Provincial Pancreatitis Center and West China-Liverpool Biomedical Research Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
11
|
M Onorato A, Fiore E, Bayo J, Casali C, Fernandez-Tomé M, Rodríguez M, Domínguez L, Argemi J, Hidalgo F, Favre C, García M, Atorrasagasti C, Mazzolini GD. SPARC inhibition accelerates NAFLD-associated hepatocellular carcinoma development by dysregulating hepatic lipid metabolism. Liver Int 2021; 41:1677-1693. [PMID: 33641248 DOI: 10.1111/liv.14857] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/27/2021] [Accepted: 02/15/2021] [Indexed: 02/13/2023]
Abstract
BACKGROUND AND AIMS Non-alcoholic fatty liver (NAFLD) and its more serious form non-alcoholic steatohepatitis increase risk of hepatocellular carcinoma (HCC). Lipid metabolic alterations and its role in HCC development remain unclear. SPARC (Secreted Protein, Acidic and Rich in Cysteine) is involved in lipid metabolism, NAFLD and diabetes, but the effects on hepatic lipid metabolism and HCC development is unknown. The aim of this study was to evaluate the role of SPARC in HCC development in the context of NAFLD. METHODS Primary hepatocyte cultures from knockout (SPARC-/- ) or wild-type (SPARC+/+ ) mice, and HepG2 cells were used to assess the effects of free fatty acids on lipid accumulation, expression of lipogenic genes and de novo triglyceride (TG) synthesis. A NAFLD-HCC model was stabilized on SPARC-/- or SPARC+/+ mice. Correlations among SPARC, lipid metabolism-related gene expression patterns and clinical prognosis were studied using HCC gene expression dataset. RESULTS SPARC-/- mice increases hepatic lipid deposits over time. Hepatocytes from SPARC-/- mice or inhibition of SPARC by an antisense adenovirus in HepG2 cells resulted in increased TG deposit, expression of lipid-related genes and nuclear translocation of SREBP1c. Human HCC database analysis revealed that SPARC negatively correlated with genes involved in lipid metabolism, and with poor survival. In NAFLD-HCC murine model, the absence of SPARC accelerates HCC development. RNA-seq study revealed that pathways related to lipid metabolism, cellular detoxification and proliferation were upregulated in SPARC-/- tumour-bearing mice. CONCLUSIONS The absence of SPARC is associated with an altered hepatic lipid metabolism, and an accelerated NAFLD-related HCC development.
Collapse
Affiliation(s)
- Agostina M Onorato
- Gene Therapy Laboratory, Instituto de Investigaciones en Medicina Traslacional, Facultad de Ciencias Biomédicas, CONICET- Universidad Austral, Buenos Aires, Argentina
| | - Esteban Fiore
- Gene Therapy Laboratory, Instituto de Investigaciones en Medicina Traslacional, Facultad de Ciencias Biomédicas, CONICET- Universidad Austral, Buenos Aires, Argentina
| | - Juan Bayo
- Gene Therapy Laboratory, Instituto de Investigaciones en Medicina Traslacional, Facultad de Ciencias Biomédicas, CONICET- Universidad Austral, Buenos Aires, Argentina
| | - Cecilia Casali
- Departamento de Ciencias Biológicas, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Cátedra de Biología Celular y Molecular, Buenos Aires, Argentina.,Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Química y Fisicoquímica Biológicas Prof. Dr. Alejandro C. Paladini (IQUIFIB), Buenos Aires, Argentina
| | - María Fernandez-Tomé
- Departamento de Ciencias Biológicas, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Cátedra de Biología Celular y Molecular, Buenos Aires, Argentina.,Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Química y Fisicoquímica Biológicas Prof. Dr. Alejandro C. Paladini (IQUIFIB), Buenos Aires, Argentina
| | - Marcelo Rodríguez
- Gene Therapy Laboratory, Instituto de Investigaciones en Medicina Traslacional, Facultad de Ciencias Biomédicas, CONICET- Universidad Austral, Buenos Aires, Argentina
| | - Luciana Domínguez
- Gene Therapy Laboratory, Instituto de Investigaciones en Medicina Traslacional, Facultad de Ciencias Biomédicas, CONICET- Universidad Austral, Buenos Aires, Argentina
| | - Josepmaría Argemi
- Josepmaria Argemi, CIMA and Clinica Universidad de Navarra, Pamplona, Spain
| | - Florencia Hidalgo
- Institute of Experimental Physiology, CONICET, School of Biochemical Sciences, University of Rosario, Rosario, Argentina
| | - Cristian Favre
- Institute of Experimental Physiology, CONICET, School of Biochemical Sciences, University of Rosario, Rosario, Argentina
| | - Mariana García
- Gene Therapy Laboratory, Instituto de Investigaciones en Medicina Traslacional, Facultad de Ciencias Biomédicas, CONICET- Universidad Austral, Buenos Aires, Argentina
| | - Catalina Atorrasagasti
- Gene Therapy Laboratory, Instituto de Investigaciones en Medicina Traslacional, Facultad de Ciencias Biomédicas, CONICET- Universidad Austral, Buenos Aires, Argentina
| | - Guillermo D Mazzolini
- Gene Therapy Laboratory, Instituto de Investigaciones en Medicina Traslacional, Facultad de Ciencias Biomédicas, CONICET- Universidad Austral, Buenos Aires, Argentina.,Liver Unit, Hospital Universitario Austral, Universidad Austral, Buenos Aires, Argentina
| |
Collapse
|
12
|
Fonseca-Camarillo G, Furuzawa-Carballeda J, Razo-López N, Barreto-Zúñiga R, Martínez-Benítez B, Yamamoto-Furusho JK. Intestinal production of secreted protein acidic and rich in cysteine (SPARC) in patients with ulcerative colitis. Immunobiology 2021; 226:152095. [PMID: 34000572 DOI: 10.1016/j.imbio.2021.152095] [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: 11/18/2020] [Revised: 03/03/2021] [Accepted: 03/28/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Ulcerative colitis (UC) is an inflammatory disease of the intestine. The genetics factors play an important role in the pathogenesis of UC. SPARC exacerbates colonic inflammatory symptoms in dextran sodium sulphate-induced murine colitis. The aim of the study was to measure the gene expression and intestinal production of SPARC in patients with UC and controls as well as, to determine its correlation with histological activity. METHODS We included 40 patients with confirmed diagnosis of UC, and 20 controls without endoscopic evidence of any type of colitis or neoplasia. The relative quantification of the gene expression was performed by real time PCR. GAPDH was used as housekeeping gene for normalization purposes and quality controls. Protein expression was determined by immunohistochemistry. RESULTS The gene expression of SPARC was increased in patients with active UC vs in remission UC and vs. controls (P = 0.005). There was no significant difference between patients with remission UC and controls. The overexpression of SPARC in patients with active UC correlated significantly with mild histological activity (P = 0.06, OR = 7.77, IC = 0.77-77.9) moderate (P = 0.06, OR = 8.1, IC 95%=0.79-82.73), and severe (P = 0.03, OR = 6.5, IC 95%=1.09-38.6). Double positive SPARC+/CD16+ cells were localized mainly in submucosa, muscular layer, and adventitia, and in perivascular inflammatory infiltrates in patients with active UC. CONCLUSION The gene and protein expression of SPARC is increased in active UC. SPARC could be a marker of intestinal inflammation and its expression correlates with histological activity.
Collapse
Affiliation(s)
- Gabriela Fonseca-Camarillo
- Inflammatory Bowel Disease Clinic. Department of Gastroenterology. Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán. México, CDMX, Mexico
| | - Janette Furuzawa-Carballeda
- Department of Immunology and Rheumatology. Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán. Mexico, CDMX, Mexico
| | - Natalia Razo-López
- Inflammatory Bowel Disease Clinic. Department of Gastroenterology. Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán. México, CDMX, Mexico
| | - Rafael Barreto-Zúñiga
- Department of Endoscopy. Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán. Mexico, CDMX, Mexico
| | - Braulio Martínez-Benítez
- Department of Pathology. Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán. Mexico, CDMX, Mexico
| | - Jesús K Yamamoto-Furusho
- Inflammatory Bowel Disease Clinic. Department of Gastroenterology. Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán. México, CDMX, Mexico.
| |
Collapse
|
13
|
McQuitty CE, Williams R, Chokshi S, Urbani L. Immunomodulatory Role of the Extracellular Matrix Within the Liver Disease Microenvironment. Front Immunol 2020; 11:574276. [PMID: 33262757 PMCID: PMC7686550 DOI: 10.3389/fimmu.2020.574276] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 10/14/2020] [Indexed: 12/12/2022] Open
Abstract
Chronic liver disease when accompanied by underlying fibrosis, is characterized by an accumulation of extracellular matrix (ECM) proteins and chronic inflammation. Although traditionally considered as a passive and largely architectural structure, the ECM is now being recognized as a source of potent damage-associated molecular pattern (DAMP)s with immune-active peptides and domains. In parallel, the ECM anchors a range of cytokines, chemokines and growth factors, all of which are capable of modulating immune responses. A growing body of evidence shows that ECM proteins themselves are capable of modulating immunity either directly via ligation with immune cell receptors including integrins and TLRs, or indirectly through release of immunoactive molecules such as cytokines which are stored within the ECM structure. Notably, ECM deposition and remodeling during injury and fibrosis can result in release or formation of ECM-DAMPs within the tissue, which can promote local inflammatory immune response and chemotactic immune cell recruitment and inflammation. It is well described that the ECM and immune response are interlinked and mutually participate in driving fibrosis, although their precise interactions in the context of chronic liver disease are poorly understood. This review aims to describe the known pro-/anti-inflammatory and fibrogenic properties of ECM proteins and DAMPs, with particular reference to the immunomodulatory properties of the ECM in the context of chronic liver disease. Finally, we discuss the importance of developing novel biotechnological platforms based on decellularized ECM-scaffolds, which provide opportunities to directly explore liver ECM-immune cell interactions in greater detail.
Collapse
Affiliation(s)
- Claire E. McQuitty
- Institute of Hepatology, Foundation for Liver Research, London, United Kingdom
- Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| | - Roger Williams
- Institute of Hepatology, Foundation for Liver Research, London, United Kingdom
- Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| | - Shilpa Chokshi
- Institute of Hepatology, Foundation for Liver Research, London, United Kingdom
- Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| | - Luca Urbani
- Institute of Hepatology, Foundation for Liver Research, London, United Kingdom
- Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| |
Collapse
|
14
|
Chen S, Zou Q, Chen Y, Kuang X, Wu W, Guo M, Cai Y, Li Q. Regulation of SPARC family proteins in disorders of the central nervous system. Brain Res Bull 2020; 163:178-189. [DOI: 10.1016/j.brainresbull.2020.05.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 04/29/2020] [Accepted: 05/05/2020] [Indexed: 12/14/2022]
|
15
|
Du Z, Lin Z, Wang Z, Liu D, Tian D, Xia L. SPOCK1 overexpression induced by platelet-derived growth factor-BB promotes hepatic stellate cell activation and liver fibrosis through the integrin α5β1/PI3K/Akt signaling pathway. J Transl Med 2020; 100:1042-1056. [PMID: 32291390 DOI: 10.1038/s41374-020-0425-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 03/16/2020] [Accepted: 03/20/2020] [Indexed: 12/13/2022] Open
Abstract
Sparc/osteonectin, cwcv, and kazal-like domain proteoglycan 1 (SPOCK1) is a matricellular protein which regulates cell proliferation, invasion, and survival but the function of SPOCK1 in liver fibrosis is obscure. In this study, we found that SPOCK1 expression increased significantly in fibrotic liver tissues and activated primary rat hepatic stellate cells (R-HSCs). SPOCK1 co-localized with α-smooth muscle actin (α-SMA) in the cytoplasm. Mechanistically, we found platelet-derived growth factor-BB (PDGF-BB) induced SPOCK1 expression by activating the PI3K/Akt/forkhead box M1 (FoxM1) signaling pathway. Intracellular SPOCK1 downregulation decreased the HSC activation, proliferation, and migration induced by PDGF-BB. Furthermore, intracellular SPOCK1 overexpression or recombinant SPOCK1 treatment promoted HSC activation, proliferation, and migration by activating the PI3K/Akt signaling pathway. Co-immunoprecipitation, double immunofluorescence staining indicated that SPOCK1 interacted with integrin α5β1, and neutralization of integrin α5β1 significantly reduced the role of recombinant SPOCK1 in HSCs. In vivo HSC-specific SPOCK1 knockdown following lentivirus administration dramatically ameliorated thioacetamide (TAA)-induced collagen deposition in rat livers. Collectively, our study indicates that SPOCK1 is crucial for hepatic fibrosis and it might be a promising therapeutic target.
Collapse
Affiliation(s)
- Zhipeng Du
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China.,Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Zhuoying Lin
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China.,Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Zhihui Wang
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China.,Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Danfei Liu
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China.,Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Dean Tian
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China. .,Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China.
| | - Limin Xia
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China. .,Institute of Liver and Gastrointestinal Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China.
| |
Collapse
|
16
|
Ghanemi A, Yoshioka M, St-Amand J. Secreted protein acidic and rich in cysteine and inflammation: Another homeostatic property? Cytokine 2020; 133:155179. [PMID: 32619797 DOI: 10.1016/j.cyto.2020.155179] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 06/12/2020] [Accepted: 06/13/2020] [Indexed: 02/08/2023]
Affiliation(s)
- Abdelaziz Ghanemi
- Functional Genomics Laboratory, Endocrinology and Nephrology Axis, CHU de Québec-Université Laval Research Center, Québec, Québec G1V 4G2, Canada; Department of Molecular Medicine, Faculty of Medicine, Laval University, Québec, Québec G1V 0A6, Canada
| | - Mayumi Yoshioka
- Functional Genomics Laboratory, Endocrinology and Nephrology Axis, CHU de Québec-Université Laval Research Center, Québec, Québec G1V 4G2, Canada
| | - Jonny St-Amand
- Functional Genomics Laboratory, Endocrinology and Nephrology Axis, CHU de Québec-Université Laval Research Center, Québec, Québec G1V 4G2, Canada; Department of Molecular Medicine, Faculty of Medicine, Laval University, Québec, Québec G1V 0A6, Canada.
| |
Collapse
|
17
|
Mukherjee S, Choi MJ, Kim SW, Yun JW. Secreted protein acidic and rich in cysteine (SPARC) regulates thermogenesis in white and brown adipocytes. Mol Cell Endocrinol 2020; 506:110757. [PMID: 32057945 DOI: 10.1016/j.mce.2020.110757] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 02/07/2020] [Accepted: 02/08/2020] [Indexed: 02/06/2023]
Abstract
SPARC, also known as osteonectin, is well known for its physiological roles in bone formation and tissue remodeling, as well as in cancer pathology; however, evidence regarding its function in adipocytes is lacking. The present study explored the physiological role of SPARC in cultured 3T3-L1 white and HIB1B brown adipocytes of murine cell lines. Treatment of recombinant SPARC upregulated the fat browning marker proteins and genes in white adipocytes and activated brown adipocytes. Conversely, knockdown of Sparc markedly reduced these genes and proteins in both cell lines. In addition, recombinant SPARC inhibited expression of adipogenic and lipogenic proteins but elevated lipolytic and fatty acid oxidation proteins. Furthermore, in silico analysis revealed that SPARC directly interacted and regulated VEGF in adipocytes. In conclusion, SPARC acts as a regulatory protein in both white and brown adipocytes by controlling thermogenesis and is thus regarded as a possible therapeutic target for treatment of obesity.
Collapse
Affiliation(s)
- Sulagna Mukherjee
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk, 38453, Republic of Korea
| | - Min Ji Choi
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk, 38453, Republic of Korea
| | - Sang Woo Kim
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung-si, Gangwon-do, 25601, Republic of Korea
| | - Jong Won Yun
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk, 38453, Republic of Korea.
| |
Collapse
|
18
|
Colak D, Al-Harazi O, Mustafa OM, Meng F, Assiri AM, Dhar DK, Broering DC. RNA-Seq transcriptome profiling in three liver regeneration models in rats: comparative analysis of partial hepatectomy, ALLPS, and PVL. Sci Rep 2020; 10:5213. [PMID: 32251301 PMCID: PMC7089998 DOI: 10.1038/s41598-020-61826-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 02/28/2020] [Indexed: 12/13/2022] Open
Abstract
The liver is a unique organ that has a phenomenal capacity to regenerate after injury. Different surgical procedures, including partial hepatectomy (PH), intraoperative portal vein ligation (PVL), and associated liver partition and portal vein ligation for staged hepatectomy (ALPPS) show clinically distinct recovery patterns and regeneration. The observable clinical differences likely mirror some underlying variations in the patterns of gene activation and regeneration pathways. In this study, we provided a comprehensive comparative transcriptomic analysis of gene regulation in regenerating rat livers temporally spaced at 24 h and 96 h after PH, PVL, and ALPPS. The time-dependent factors appear to be the most important determinant of post-injury alterations of gene expression in liver regeneration. Gene expression profile after ALPPS showed more similar expression pattern to the PH than the PVL at the early phase of the regeneration. Early transcriptomic changes and predicted upstream regulators that were found in all three procedures included cell cycle associated genes (E2F1, CCND1, FOXM1, TP53, and RB1), transcription factors (Myc, E2F1, TBX2, FOXM1), DNA replication regulators (CDKN1A, EZH2, RRM2), G1/S-transition regulators (CCNB1, CCND1, RABL6), cytokines and growth factors (CSF2, IL-6, TNF, HGF, VEGF, and EGF), ATM and p53 signaling pathways. The functional pathway, upstream, and network analyses revealed both unique and overlapping molecular mechanisms and pathways for each surgical procedure. Identification of molecular signatures and regenerative signaling pathways for each surgical procedure further our understanding of key regulators of liver regeneration as well as patient populations that are likely to benefit from each procedure.
Collapse
Affiliation(s)
- Dilek Colak
- Biostatistics, Epidemiology, and Scientific Computing Department, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia.
| | - Olfat Al-Harazi
- Biostatistics, Epidemiology, and Scientific Computing Department, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Osama M Mustafa
- Biostatistics, Epidemiology, and Scientific Computing Department, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Fanwei Meng
- Department of Surgery and Organ Transplantation Center, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
- Comparative Medicine Department, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Abdullah M Assiri
- Comparative Medicine Department, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
- Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
- College of Medicine, AlFaisal University, Riyadh, Saudi Arabia
| | - Dipok K Dhar
- Department of Surgery and Organ Transplantation Center, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia.
- Comparative Medicine Department, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia.
- Institute for Liver and Digestive Health, University College London, Royal Free Hospital, London, UK.
| | - Dieter C Broering
- Department of Surgery and Organ Transplantation Center, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
- College of Medicine, AlFaisal University, Riyadh, Saudi Arabia
| |
Collapse
|
19
|
Akaiwa M, Fukui E, Matsumoto H. Tubulointerstitial nephritis antigen-like 1 deficiency alleviates age-dependent depressed ovulation associated with ovarian collagen deposition in mice. Reprod Med Biol 2020; 19:50-57. [PMID: 31956285 PMCID: PMC6955583 DOI: 10.1002/rmb2.12301] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 08/16/2019] [Accepted: 09/01/2019] [Indexed: 12/14/2022] Open
Abstract
PURPOSE This study aimed to examine whether the Tinagl1 might be associated with ovulation in aged females and reproductive age-associated fibrosis in the stroma of the ovary. METHODS To address the ovulatory ability and quality of ovulated oocytes, we induced ovulation by treatment with equine chorionic gonadotropin (eCG) and human chorionic gonadotropin (hCG) followed by in vitro fertilization. We also performed Picrosirius Red (PSR) staining to evaluate ovarian collagen deposition. RESULTS As compared to ovulation in 8- to 9-month-old Tinagl1flox/flox mice, the number of ovulated oocytes from Tinagl1flox/flox mice decreased in an age-dependent manner in mice more than 10-11 months old, whereas the ovulated oocyte numbers in Tinagl1 -/- mice decreased significantly at 14-15 months. In vitro fertilization followed by embryo culture demonstrated the normal developmental potential of Tinagl1-null embryos during the preimplantation period. PSR staining indicated that collagen was found throughout the ovarian stroma in an age-dependent manner in Tinagl1flox/flox females, whereas those distributions were delayed to 14-15 months in Tinagl1 -/- females. This timing was consistent with the delayed timing of age-related decline of ovulation in Tinagl1 -/- females. CONCLUSIONS The alleviation of age-associated depression of ovulation was caused by delayed ovarian collagen deposition in Tinagl1-null female mice.
Collapse
Affiliation(s)
- Masato Akaiwa
- Laboratory of Animal Breeding and ReproductionDivision of Animal ScienceSchool of AgricultureUtsunomiya UniversityUtsunomiyaJapan
| | - Emiko Fukui
- Laboratory of Animal Breeding and ReproductionDivision of Animal ScienceSchool of AgricultureUtsunomiya UniversityUtsunomiyaJapan
- Center for Bioscience Research and EducationUtsunomiya UniversityUtsunomiyaJapan
| | - Hiromichi Matsumoto
- Laboratory of Animal Breeding and ReproductionDivision of Animal ScienceSchool of AgricultureUtsunomiya UniversityUtsunomiyaJapan
- Center for Bioscience Research and EducationUtsunomiya UniversityUtsunomiyaJapan
| |
Collapse
|
20
|
Wang J, Ding Y, Zhou W. Albumin self-modified liposomes for hepatic fibrosis therapy via SPARC-dependent pathways. Int J Pharm 2019; 574:118940. [PMID: 31830578 DOI: 10.1016/j.ijpharm.2019.118940] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 11/17/2019] [Accepted: 12/06/2019] [Indexed: 12/16/2022]
Abstract
Activated hepatic stellate cells (HSCs) have a central role in the progression of liver fibrosis and express a large amount of secreted protein, acidic and rich in cysteine (SPARC), a specific protein-binding protein. In this study, we reported the preparation and evaluation of naringenin (Nar) -loaded albumin self-modified liposomes (NaAlLs), which delivered Nar, a specific Smad3 inhibitor that blocked the TGF-β/Smad3 signaling pathway and played an anti-fibrosis role. After a series of characterization, it was found that NaAlLs had favorable dispersion (PDI < 0.15) with an average particle size of about 120 nm and high entrapment efficiency (>85%), albumin coated the surface of liposomes or embedded in phospholipid bilayer by interaction with the encapsulated naringenin and phospholipid molecules during the preparation of liposomes. The amount of albumin modified to the surface of NaAlLs by this method is not only more than that of the physical adsorption method, but also the binding force between albumin and liposomes is stronger. The albumin modified to the surface of NaAlLs greatly reduced the aggregation of liposomes and drug leakage and increased the stability of liposomes. More importantly, the uptake of NaAlLs by activated HSCs was 1.5 times higher than that of Nar-loaded liposomes (NaLs), suggesting that NaAlLs specifically increased targeting of activated HSCs via albumin and SPARC-dependent pathways. As expected, NaAlLs was more effective in improving liver fibrosis than the NaLs or the inclusion complex solution of Nar and Hydroxypropyl-β-cyclodextrin (NaICS). The results suggested that NaAlLs was a promising drug delivery system, which could target drug delivery to activated HSC for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Jianzhu Wang
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, Jiangsu 210009, China; School of Pharmaceutical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong 271016, China
| | - Yu Ding
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | - Wei Zhou
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, Jiangsu 210009, China.
| |
Collapse
|
21
|
Li T, Tan X, Zhu S, Zhong W, Huang B, Sun J, Li F, Wang Y. SPARC induces phenotypic modulation of human brain vascular smooth muscle cells via AMPK/mTOR-mediated autophagy. Neurosci Lett 2019; 712:134485. [DOI: 10.1016/j.neulet.2019.134485] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 08/15/2019] [Accepted: 09/05/2019] [Indexed: 12/24/2022]
|
22
|
Omi S, Yamanouchi K, Nakamura K, Matsuwaki T, Nishihara M. Reduced fibrillar collagen accumulation in skeletal muscle of secreted protein acidic and rich in cysteine (SPARC)-null mice. J Vet Med Sci 2019; 81:1649-1654. [PMID: 31582603 PMCID: PMC6895640 DOI: 10.1292/jvms.19-0485] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
We have previously shown that secreted protein acidic and rich in cysteine (SPARC) promotes myogenic differentiation of rat skeletal muscle progenitor cells in vitro, and in vivo small interfering RNA (siRNA)-mediated transient suppression of SPARC expression in skeletal muscle of mice causes atrophic changes of myofibers, suggesting that SPARC plays a role in the maintenance of skeletal muscle function. In order to know the effect of long-term deficiency of SPARC on skeletal muscle, we performed phenotypic analyses of skeletal muscle of SPARC-null mice. Age-associated changes of myofiber diameters were comparable between wild type (WT) and SPARC-null mice at all ages examined, indicating that the growth of myofibers is unaffected by the absence of SPARC. On the other hand, accumulation of fibrillar collagen was significantly reduced in SPARC-null mice compared to WT mice after 5 months of age without significant changes of collagen I gene expression. The results obtained in the present study suggest that SPARC plays a role to maintain the stiffness of skeletal muscle by regulating collagen accumulation.
Collapse
Affiliation(s)
- Sanae Omi
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Keitaro Yamanouchi
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Katsuyuki Nakamura
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Takashi Matsuwaki
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Masugi Nishihara
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| |
Collapse
|
23
|
Peixoto E, Atorrasagasti C, Malvicini M, Fiore E, Rodriguez M, Garcia M, Finocchieto P, Poderoso JJ, Corrales F, Mazzolini G. SPARC gene deletion protects against toxic liver injury and is associated to an enhanced proliferative capacity and reduced oxidative stress response. Oncotarget 2019; 10:4169-4179. [PMID: 31289615 PMCID: PMC6609249 DOI: 10.18632/oncotarget.9456] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 03/31/2016] [Indexed: 12/27/2022] Open
Abstract
SPARC, also known as osteonectin and BM-40, is a matricellular protein with a number of biological functions. Hepatic SPARC expression is induced in response to thioacetamide, bile-duct ligation, and acute injuries such as concanavalin A and lipopolysacharide (LPS)/D-galactosamine. We have previously demonstrated that the therapeutic inhibition of SPARC or SPARC gene deletion protected mice against liver injury. We investigated the mechanisms involved in the protective effect of SPARC inhibition in mice. We performed a proteome analysis of livers from SPARC+/+ and SPARC-/- mice chronically treated with thioacetamide. Catalase activity, carbonylation levels, oxidative stress response, and mitochondrial function were studied. Genomic analysis revealed that SPARC-/- mice had an increased expression of cell proliferation genes. Proteins involved in detoxification of reactive oxygen species such as catalase, peroxirredoxine-1, and glutathione-S-transferase P1 and Mu1 were highly expressed as evidenced by proteome analysis; hepatic catalase activity was increased in SPARC-/- mice. Oxidative stress response and carbonylation levels were lower in livers from SPARC-/- mice. Hepatic mitochondria showed lower levels of nitrogen reactive species in the SPARC-/- concanavalin A-treated mice. Mitochondrial morphology was preserved, and its complex activity reduced in SPARC-/- mice. In conclusion, our data suggest that the protection associated with SPARC gene deletion may be partially due to a higher proliferative capacity of hepatocytes and an enhanced oxidative stress defense in SPARC-/- mice after liver injury.
Collapse
Affiliation(s)
- Estanislao Peixoto
- Gene Therapy Laboratory, Instituto de Investigaciones Médicas Aplicadas-CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Buenos Aires, Argentina
| | - Catalina Atorrasagasti
- Gene Therapy Laboratory, Instituto de Investigaciones Médicas Aplicadas-CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Buenos Aires, Argentina
| | - Mariana Malvicini
- Gene Therapy Laboratory, Instituto de Investigaciones Médicas Aplicadas-CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Buenos Aires, Argentina
| | - Esteban Fiore
- Gene Therapy Laboratory, Instituto de Investigaciones Médicas Aplicadas-CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Buenos Aires, Argentina
| | - Marcelo Rodriguez
- Gene Therapy Laboratory, Instituto de Investigaciones Médicas Aplicadas-CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Buenos Aires, Argentina
| | - Mariana Garcia
- Gene Therapy Laboratory, Instituto de Investigaciones Médicas Aplicadas-CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Buenos Aires, Argentina
| | | | | | | | - Guillermo Mazzolini
- Gene Therapy Laboratory, Instituto de Investigaciones Médicas Aplicadas-CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Buenos Aires, Argentina
| |
Collapse
|
24
|
Sun W, Feng J, Yi Q, Xu X, Chen Y, Tang L. SPARC acts as a mediator of TGF-β1 in promoting epithelial-to-mesenchymal transition in A549 and H1299 lung cancer cells. Biofactors 2018; 44:453-464. [PMID: 30346081 DOI: 10.1002/biof.1442] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 05/25/2018] [Indexed: 12/14/2022]
Abstract
Migration and metastasis of tumor cells greatly contributes to the failure of cancer treatment. Recently, the extracellular protein secreted protein acidic and rich in cysteine (SPARC) has been reported closely related to tumorigenesis. Some articles have suggested that SPARC promoted metastasis in several highly metastatic tumors. However, there are also some studies shown that SPARC acted as an antitumor factor. SPARC-induced epithelial-to-mesenchymal transition (EMT) in melanoma cells and promoted EMT in hepatocellular carcinoma. Therefore, the role of SPARC in tumorigenesis and its relationship with EMT is still unclear. In this study, we investigated the expression change of SPARC in A549 and H1299 lung cancer cells undergoing EMT process. Our study indicated that SPARC was upregulated in A549 and H1299 cells EMT process. We further investigated the function of SPARC on proliferation, migration, and EMT process of A549 and H1299 cells. Overexpression of SPARC promoted the migration and EMT of A549 and H1299 cells. Knockdown SPARC inhibited the EMT of A549 cells. Overexpression of SPARC induced the increased expression of p-Akt and P-ERK. Furthermore, exogenous SPARC peptide promoted transforming growth factor (TGF)-β1-induced EMT of A549 and H1299 cells. SPARC knockdown partially eliminated TGF-β1 function in inducing EMT of A549 cells. SPARC follistatin-like functional domain reduced the expression of E-cadherin, but had no effect on the expression of p-Akt and p-ERK. In conclusion, we elucidated that SPARC contributes to tumorigenesis by promoting migration and EMT of A549 and H1299 lung cancer cells. These results will provide some new suggestion for lung cancer treatment. © 2018 BioFactors, 44(5):453-464, 2018.
Collapse
Affiliation(s)
- Weichao Sun
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Jianguo Feng
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Qian Yi
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
- Department of Physiology, College of Preclinical Medicine, Southwest Medical University, Luzhou, Sichuan Province, Luzhou, Sichuan Province, China
| | - Xichao Xu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Ying Chen
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Liling Tang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| |
Collapse
|
25
|
Tanaka M, Takagi T, Naito Y, Uchiyama K, Hotta Y, Toyokawa Y, Ushiroda C, Hirai Y, Aoi W, Higashimura Y, Mizushima K, Okayama T, Katada K, Kamada K, Ishikawa T, Handa O, Itoh Y. Secreted protein acidic and rich in cysteine functions in colitis via IL17A regulation in mucosal CD4 + T cells. J Gastroenterol Hepatol 2018; 33:671-680. [PMID: 28582593 DOI: 10.1111/jgh.13842] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Revised: 05/29/2017] [Accepted: 06/01/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIM Secreted protein acidic and rich in cysteine (SPARC) is a matricellular glycol that regulates cell proliferation, tissue repair, and tumorigenesis. Despite evidence linking SPARC to inflammation, the mechanisms are unclear. Accordingly, the role of SPARC in intestinal inflammation was investigated. METHODS Colitis was induced in wild-type (WT) and SPARC knockout (KO) mice using trinitrobenzene sulfonic acid (TNBS). Colons were assessed for damage; leukocyte infiltration; Tnf, Ifng, Il17a, and Il10 mRNA expression; and histology. Cytokine profiling of colonic lamina propria mononuclear cells (LPMCs) was performed by flow cytometry. Naïve CD4+ T cells were isolated from WT and SPARC KO mouse spleens, and the effect of SPARC on Th17 cell differentiation was examined. Recombination activating gene 1 knockout (RAG1 KO) mice reconstituted with T cells from either WT or SPARC KO mice were investigated. RESULTS Trinitrobenzene sulfonic acid exposure significantly reduced bodyweight and increased mucosal inflammation, leukocyte infiltration, and Il17a mRNA expression in WT relative to SPARC KO mice. The percentage of IL17A-producing CD4+ T cells among LPMCs from KO mice was lower than that in WT mice when both groups were exposed to TNBS. Th17 cell differentiation was suppressed in cells from SPARC KO mice. In the T cell transfer colitis model, RAG1 KO mice receiving T cells from WT mice were more severely affected than those reconstituted with cells from SPARC KO mice. CONCLUSIONS Secreted protein acidic and rich in cysteine accelerates colonic mucosal inflammation via modulation of IL17A-producing CD4+ T cells. SPARC is a potential therapeutic target for conditions involving intestinal inflammation.
Collapse
Affiliation(s)
- Makoto Tanaka
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tomohisa Takagi
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yuji Naito
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kazuhiko Uchiyama
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yuma Hotta
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yuki Toyokawa
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Chihiro Ushiroda
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yasuko Hirai
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Wataru Aoi
- Laboratory of Health Science, Kyoto Prefectural University, Kyoto, Japan
| | - Yasuki Higashimura
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Katsura Mizushima
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tetsuya Okayama
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kazuhiro Katada
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kazuhiro Kamada
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takeshi Ishikawa
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Osamu Handa
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yoshito Itoh
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
26
|
SPARC expression is associated with hepatic injury in rodents and humans with non-alcoholic fatty liver disease. Sci Rep 2018; 8:725. [PMID: 29335425 PMCID: PMC5768809 DOI: 10.1038/s41598-017-18981-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 12/15/2017] [Indexed: 12/29/2022] Open
Abstract
Mechanisms that control progression from simple steatosis to steato-hepatitis and fibrosis in patients with non-alcoholic fatty liver disease (NAFLD) are unknown. SPARC, a secreted matricellular protein, is over-expressed in the liver under chronic injury. Contribution of SPARC accumulation to disease severity is largely unknown in NAFLD. We assessed the hypothesis that SPARC is increased in livers with more necrosis and inflammation and could be associated with more fibrosis. qrt-PCR, immunohistochemistry, and ELISA were employed to localize and quantify changes in SPARC in 62 morbidly obese patients with NAFLD/NASH and in a mouse model of diet-induced-NASH. Results were correlated with the severity of NAFLD/NASH. In obese patients 2 subgroups were identified with either high SPARC expression (n = 16) or low SPARC expression (n = 46) in the liver, with a cutoff of 1.2 fold expression. High expression of SPARC paralleled hepatocellular damage and increased mRNA expression of pro-fibrogenic factors in the liver. In line with these findings, in the NASH animal model SPARC knockout mice were protected from inflammatory injury, and showed less inflammation and fibrosis. Hepatic SPARC expression is associated with liver injury and fibrogenic processes in NAFLD. SPARC has potential as preventive or therapeutic target in NAFLD patients.
Collapse
|
27
|
Aseer KR, Silvester AJ, Kumar A, Choi MS, Yun JW. SPARC paucity alleviates superoxide-mediated oxidative stress, apoptosis, and autophagy in diabetogenic hepatocytes. Free Radic Biol Med 2017; 108:874-895. [PMID: 28499910 DOI: 10.1016/j.freeradbiomed.2017.05.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 05/05/2017] [Accepted: 05/08/2017] [Indexed: 12/14/2022]
Abstract
Secreted protein acidic and rich in cysteine (SPARC) is known to play a previously unappreciated role in diabetes, but its precise mechanism in liver/hepatocyte pathology remains unknown. Inhibition of SPARC is critical in resolving candidate pathogenic events such as production of reactive oxygen species (ROS), which are broadly considered for their roles in diabetes, and is capable of protecting functional hepatocytes. Here, we provide in vitro and in vivo evidence demonstrating pathological correlations between SPARC and streptozotocin (STZ)-induced diabetic rat livers as well as cultured hepatocytes induced by diabetogenic stimuli. Under these conditions, transient SPARC silencing was carried out to investigate the role of SPARC in the pathogenesis of pro-diabetic hepatocyte damage and dysfunction. The constitutive expression of SPARC in hepatocytes was up-regulated under a diabetic environment. In addition, Nox4-dependent superoxide generation contributed to increased expression of SPARC, and this was inhibited by tiron and pharmacological or genetic inactivation of Nox4-containing NADPH oxidase. Remarkably, SPARC deficiency inhibited diabetic stimuli-induced elevation of superoxide production and resolved salient features of hepatocyte damage such as impaired cytoprotection, inflammation, apoptosis, and autophagy. At the same time, links between SPARC, integrin-β1, Nox4-derived superoxide, and JNK signaling provide a basis for these phenotypes. Taken together with the observations that SPARC deficiency had protective effects on hepatocytes via a favorable inhibition profile, functional knowledge of SPARC may offer a unique therapeutic approach to preserve hepatocellular fate decisions in diabetes.
Collapse
Affiliation(s)
- Kanikkai Raja Aseer
- Department of Biotechnology, Daegu University, Kyungsan, Kyungbuk 712-714, Republic of Korea
| | | | - Anuj Kumar
- Bioinformatics and Documentation Laboratory, Uttarakhand Council for Biotechnology, Dehradun 248007, India
| | - Myung-Sook Choi
- Department of Food Science and Nutrition & Center for Food and Nutritional Genomics Research, Kyungpook National University, Daegu 702-701, Republic of Korea
| | - Jong Won Yun
- Department of Biotechnology, Daegu University, Kyungsan, Kyungbuk 712-714, Republic of Korea.
| |
Collapse
|
28
|
Jørgensen LH, Jepsen PL, Boysen A, Dalgaard LB, Hvid LG, Ørtenblad N, Ravn D, Sellathurai J, Møller-Jensen J, Lochmüller H, Schrøder HD. SPARC Interacts with Actin in Skeletal Muscle in Vitro and in Vivo. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:457-474. [DOI: 10.1016/j.ajpath.2016.10.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 09/30/2016] [Accepted: 10/18/2016] [Indexed: 01/06/2023]
|
29
|
Liu J, Li M, Gong J, Han P, Wang Y, Li D, Tian D, Liao J. Knockdown of histidine-rich calcium-binding protein (HRC) suppresses liver fibrosis by inhibiting the activation of hepatic stellate cells. Biol Open 2017; 6:29-34. [PMID: 27881436 PMCID: PMC5278420 DOI: 10.1242/bio.019828] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The histidine-rich calcium-binding protein (HRC) is a regulator of Ca2+ homeostasis and it plays a significant role in hepatocellular carcinoma (HCC) progression. However, the relationship between HRC and liver fibrogenesis is still unknown. Our data demonstrates that HRC was upregulated in fibrotic liver and activated hepatic stellate cells (HSCs). TGF-β treatment increased α-SMA and HRC expression dose-dependently in HSCs. Repression of HRC reduced α-SMA, CTGF and collagen expression, and inhibited HSC proliferation and migration. In addition, we found that the anti-fibrosis effect of HRC knockdown was associated with endoplasmic reticulum (ER) stress. Silencing of HRC decreased the expression of ER stress and autophagy markers. Moreover, ER stress agonist thapsigargin (TG) enhanced, whereas ER stress antagonist 4-phenylbutyric acid (4-PBA) alleviated HSCs activation and autophagy. In conclusion, these data indicate that depletion of HRC inhibited HSC activation through the ER stress pathway, and HRC may be a potential regulator of liver fibrosis.
Collapse
Affiliation(s)
- Jingmei Liu
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Mengke Li
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.,Department of Gastroenterology, Zhoushan Hospital, Zhoushan 316000, China
| | - Jin Gong
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ping Han
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yunwu Wang
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Dongxiao Li
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Dean Tian
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jiazhi Liao
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
30
|
Zhao J, Peng L, Cui R, Guo X, Yan M. Dimethyl α-ketoglutarate reduces CCl 4-induced liver fibrosis through inhibition of autophagy in hepatic stellate cells. Biochem Biophys Res Commun 2016; 481:90-96. [PMID: 27823933 DOI: 10.1016/j.bbrc.2016.11.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 11/03/2016] [Indexed: 02/07/2023]
Abstract
Sustained activation of hepatic stellate cells (HSCs) leads to liver fibrosis. Autophagy fuels the activation of HSCs by generation of ATP. Our previous research demonstrated an inhibitory effect of dimethyl α-ketoglutarate (DMKG) on HSCs activation in vitro. In the current study, we demonstrated that DMKG reduced CCl4-induced liver fibrosis in Wistar rats. Then, with the use of the HSC-T6 cell lines and double immunofluorescent staining of liver sections, we showed that the anti-fibrotic effect occurred through the inhibition of the autophagy of HSCs. Both experiments showed that DMKG could inhibit autophagy and activation of HSCs, and that the activation of HSCs was down-regulated with autophagy. In addition, we showed that DMKG could lead to lipid droplet accumulation and decrease cellular ATP content in HSCs. Furthermore, the mechanism of how DMKG inhibited autophagy of HSCs was explored in vitro with the use of c646 (a competitive inhibitor of acetyl-coenzyme A which binds to the acetyltransferase EP300) and lipoic acid (an alternative acetyl-coenzyme A -replenishing agent to DMKG), and showed that both acetyl-coenzyme A and EP300 were involved. Collectively, our study investigated the possible role of DMKG in preventing liver fibrosis and HSCs activation. We showed that DMKG may be a potential therapeutic agent for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Jianjian Zhao
- Department of Geriatric Gastroenterology, Qilu Hospital of Shandong University, Jinan, 250012, China.
| | - Lei Peng
- Department of Geriatric Gastroenterology, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Ruibing Cui
- Department of Geriatric Gastroenterology, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Xiaolan Guo
- Department of Geriatric Gastroenterology, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Ming Yan
- Department of Geriatric Gastroenterology, Qilu Hospital of Shandong University, Jinan, 250012, China.
| |
Collapse
|
31
|
Mesenchymal Stem/Stromal Cells in Liver Fibrosis: Recent Findings, Old/New Caveats and Future Perspectives. Stem Cell Rev Rep 2016; 11:586-97. [PMID: 25820543 DOI: 10.1007/s12015-015-9585-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Mesenchymal stem/stromal cells (MSCs) are progenitors which share plastic-adherence capacity and cell surface markers but have different properties according to their cell and tissue sources and to culture conditions applied. Many recent publications suggest that MSCs can differentiate into hepatic-like cells, which can be a consequence of either a positive selection of rare in vivo pluripotent cells or of the original plasticity of some cells contributing to MSC cultures. A possible role of MSCs in hereditary transmission of obesity and/or diabetes as well as properties of MSCs regarding immunomodulation, cell fusion and exosome release capacities are discussed according to recent literature. Limitations in methods used to track MSCs in vivo especially in the context of liver cirrhosis are addressed as well as strategies explored to enhance their migratory, survival and proliferation properties, which are known to be relevant for their future clinical use. Current knowledge regarding mechanisms involved in liver cirrhosis amelioration mediated by naïve and genetically modified MSCs as well as the effects of applying preconditioning and combined strategies to improve their therapeutic effects are evaluated. Finally, first reports of GMP guidelines and biosafety issues in MSCs applications are discussed.
Collapse
|
32
|
Sawyer AJ, Kyriakides TR. Matricellular proteins in drug delivery: Therapeutic targets, active agents, and therapeutic localization. Adv Drug Deliv Rev 2016; 97:56-68. [PMID: 26763408 DOI: 10.1016/j.addr.2015.12.016] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 12/17/2015] [Accepted: 12/17/2015] [Indexed: 02/06/2023]
Abstract
Extracellular matrix is composed of a complex array of molecules that together provide structural and functional support to cells. These properties are mainly mediated by the activity of collagenous and elastic fibers, proteoglycans, and proteins such as fibronectin and laminin. ECM composition is tissue-specific and could include matricellular proteins whose primary role is to modulate cell-matrix interactions. In adults, matricellular proteins are primarily expressed during injury, inflammation and disease. Particularly, they are closely associated with the progression and prognosis of cardiovascular and fibrotic diseases, and cancer. This review aims to provide an overview of the potential use of matricellular proteins in drug delivery including the generation of therapeutic agents based on the properties and structures of these proteins as well as their utility as biomarkers for specific diseases.
Collapse
|
33
|
Weiskirchen R. Hepatoprotective and Anti-fibrotic Agents: It's Time to Take the Next Step. Front Pharmacol 2016; 6:303. [PMID: 26779021 PMCID: PMC4703795 DOI: 10.3389/fphar.2015.00303] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 12/11/2015] [Indexed: 12/21/2022] Open
Abstract
Hepatic fibrosis and cirrhosis cause strong human suffering and necessitate a monetary burden worldwide. Therefore, there is an urgent need for the development of therapies. Pre-clinical animal models are indispensable in the drug discovery and development of new anti-fibrotic compounds and are immensely valuable for understanding and proofing the mode of their proposed action. In fibrosis research, inbreed mice and rats are by far the most used species for testing drug efficacy. During the last decades, several hundred or even a thousand different drugs that reproducibly evolve beneficial effects on liver health in respective disease models were identified. However, there are only a few compounds (e.g., GR-MD-02, GM-CT-01) that were translated from bench to bedside. In contrast, the large number of drugs successfully tested in animal studies is repeatedly tested over and over engender findings with similar or identical outcome. This circumstance undermines the 3R (Replacement, Refinement, Reduction) principle of Russell and Burch that was introduced to minimize the suffering of laboratory animals. This ethical framework, however, represents the basis of the new animal welfare regulations in the member states of the European Union. Consequently, the legal authorities in the different countries are halted to foreclose testing of drugs in animals that were successfully tested before. This review provides a synopsis on anti-fibrotic compounds that were tested in classical rodent models. Their mode of action, potential sources and the observed beneficial effects on liver health are discussed. This review attempts to provide a reference compilation for all those involved in the testing of drugs or in the design of new clinical trials targeting hepatic fibrosis.
Collapse
Affiliation(s)
- Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy, and Clinical Chemistry, RWTH University Hospital Aachen Aachen, Germany
| |
Collapse
|
34
|
Yamanaka O, Kitano-Izutani A, Tomoyose K, Reinach PS. Pathobiology of wound healing after glaucoma filtration surgery. BMC Ophthalmol 2015; 15 Suppl 1:157. [PMID: 26818010 PMCID: PMC4895697 DOI: 10.1186/s12886-015-0134-8] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Conjunctival and subconjunctival fibrogenesis and inflammation are sight compromising side effects that can occur subsequent to glaucoma filtration surgery. Despite initial declines in intraocular pressure resulting from increasing aqueous outflow, one of the activated responses includes marshalling of proinflammatory and pro-fibrogenic cytokine mediator entrance into the aqueous through a sclerostomy window and their release by local cells, as well as infiltrating activated immune cells. These changes induce dysregulated inflammation, edema and extracellular matrix remodeling, which occlude outflow facility. A number of therapeutic approaches are being taken to offset declines in outflow facility since the current procedure of inhibiting fibrosis with either mitomycin C (MMC) or 5-fluorouracil (5-FU) injection is nonselective. One of them entails developing a new strategy for reducing fibrosis induced by wound healing responses including myofibroblast transdifferentiation and extracellular matrix remodeling in tissue surrounding surgically created shunts. The success of this endeavor is predicated on having a good understanding of conjunctival wound healing pathobiology. In this review, we discuss the roles of inappropriately activated growth factor and cytokine receptor linked signaling cascades inducing conjunctival fibrosis/scarring during post-glaucoma surgery wound healing. Such insight may identify drug targets for blocking fibrogenic signaling and excessive fibrosis which reduces rises in outflow facility resulting from glaucoma filtration surgery.
Collapse
Affiliation(s)
- Osamu Yamanaka
- Department of Ophthalmology, Wakayama Medical University, 811-1 Kimiidera, Wakayama, Wakayama, 641-0012, Japan.
| | - Ai Kitano-Izutani
- Department of Ophthalmology, Wakayama Medical University, 811-1 Kimiidera, Wakayama, Wakayama, 641-0012, Japan.
| | - Katsuo Tomoyose
- Department of Ophthalmology, Wakayama Medical University, 811-1 Kimiidera, Wakayama, Wakayama, 641-0012, Japan.
| | - Peter S Reinach
- Departments of Ophthalmology and Optometry Wenzhou Medical University, Wenzhou, 325027, People's Republic of China.
| |
Collapse
|
35
|
Steiner JL, Pruznak AM, Navaratnarajah M, Lang CH. Alcohol Differentially Alters Extracellular Matrix and Adhesion Molecule Expression in Skeletal Muscle and Heart. Alcohol Clin Exp Res 2015; 39:1330-40. [PMID: 26108259 DOI: 10.1111/acer.12771] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 05/01/2015] [Indexed: 02/06/2023]
Abstract
BACKGROUND The production of fibrosis in response to chronic alcohol abuse is well recognized in liver but has not been fully characterized in striated muscle and may contribute to functional impairment. Therefore, the purpose of this study was to use an unbiased discovery-based approach to determine the effect of chronic alcohol consumption on the expression profile of genes important for cell-cell and cell-extracellular matrix (ECM) interactions in both skeletal and cardiac muscle. METHODS Adult male rats were pair-fed an alcohol-containing liquid diet or control diet for 24 weeks, and skeletal muscle (gastrocnemius) and heart were collected in the freely fed state. A pathway-focused gene expression polymerase chain reaction array was performed on these tissues to assess mRNA content for 84 ECM proteins, and selected proteins were confirmed by Western blot analysis. RESULTS In gastrocnemius, alcohol feeding up-regulated the expression of 11 genes and down-regulated the expression of 1 gene. Alcohol increased fibrosis as indicated by increased mRNA and/or protein for collagens α1(I), α2(I), α1(III), and α2(IV) as well as hydroxyproline. Alcohol also increased α-smooth muscle actin protein, an index of myofibroblast activation, but no concomitant change in transforming growth factor-β was detected. The mRNA and protein content for other ECM components, such as integrin-α5, L-selectin, PECAM, SPARC, and ADAMTS2, were also increased by alcohol. Only laminin-α3 mRNA was decreased in gastrocnemius from alcohol-fed rats, while 66 ECM- or cell adhesion-related mRNAs were unchanged by alcohol. For heart, expression of 16 genes was up-regulated, expression of 3 genes was down-regulated, and 65 mRNAs were unchanged by alcohol; there were no common alcohol-induced gene expression changes between heart and skeletal muscle. Finally, alcohol increased tumor necrosis factor-α and interleukin (IL)-12 mRNA in both skeletal and cardiac muscle, but IL-6 mRNA was increased and IL-10 mRNA decreased only in skeletal muscle. CONCLUSIONS These data demonstrate a fibrotic response in striated muscle from chronic alcohol-fed rats which is tissue specific in nature, suggesting different regulatory mechanisms.
Collapse
Affiliation(s)
- Jennifer L Steiner
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania
| | - Anne M Pruznak
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania
| | - Maithili Navaratnarajah
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania
| | - Charles H Lang
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania
| |
Collapse
|
36
|
Strategies to prevent and reverse liver fibrosis in humans and laboratory animals. Arch Toxicol 2015; 89:1727-50. [PMID: 25963329 DOI: 10.1007/s00204-015-1525-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 04/28/2015] [Indexed: 02/07/2023]
Abstract
Liver fibrosis results from chronic damage to the liver in conjunction with various pathways and is mediated by a complex microenvironment. Based on clinical observations, it is now evident that fibrosis is a dynamic, bidirectional process with an inherent capacity for recovery and remodeling. The major mechanisms involved in liver fibrosis include the repetitive injury of hepatocytes, the activation of the inflammatory response after injury stimulation, and the activation and proliferation of hepatic stellate cells (HSCs), which represents the major extracellular matrix (ECM)-producing cells, stimulated by hepatocyte injury and inflammation. The microenvironment in the liver is synergistically regulated abnormal ECM deposition, scar formation, angiogenesis, and fibrogenesis. Moreover, recent studies have clarified novel mechanism in fibrosis such as epigenetic regulation of HSCs, the leptin and PPARγ pathways, the coagulation system, and even autophagy. Uncovering the mechanisms of liver fibrogenesis provides a basis to develop potential therapies to reverse and treat the fibrotic response, thereby improving the outcomes of patients with chronic liver disease. Although both scientific and clinical challenges remain, emerging studies attempt to reveal the ideal anti-fibrotic drug that could be easily delivered to the liver with high specificity and low toxicity. This review highlights the mechanisms, including novel pathways underlying fibrogenesis that may be translated into preventive and treatment strategies, reviews both current and novel agents that target specific pathways or multiple targets, and discusses novel drug delivery systems such as nanotechnology that can be applied in the treatment of liver fibrosis. In addition, we also discuss some current treatment strategies that are being applied in animal models and in clinical trials.
Collapse
|
37
|
Piccioni F, Fiore E, Bayo J, Atorrasagasti C, Peixoto E, Rizzo M, Malvicini M, Tirado-González I, García MG, Alaniz L, Mazzolini G. 4-methylumbelliferone inhibits hepatocellular carcinoma growth by decreasing IL-6 production and angiogenesis. Glycobiology 2015; 25:825-35. [PMID: 25882295 DOI: 10.1093/glycob/cwv023] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 04/11/2015] [Indexed: 02/07/2023] Open
Abstract
Cirrhosis is characterized by an excessive accumulation of extracellular matrix components including hyaluronic acid (HA) and is widely considered a preneoplastic condition for hepatocellular carcinoma (HCC). 4-Methylumbelliferone (4MU) is an inhibitor of HA synthesis and has anticancer activity in an orthotopic HCC model with underlying fibrosis. Our aim was to explore the effects of HA inhibition by 4MU orally administered on tumor microenvironment. Hepa129 tumor cells were inoculated orthotopically in C3H/HeJ male mice with fibrosis induced by thioacetamide. Mice were orally treated with 4MU. The effects of 4MU on angiogenesis were evaluated by immunostaining of CD31 and quantification of proangiogenic factors (vascular endothelial growth factor, VEGF, interleukin-6, IL-6 and C-X-C motif chemokine 12, CXCL12). IL-6 was also quantified in Hepa129 cells in vitro after treatment with 4MU. Migration of endothelial cells and tube formation were also analyzed. As a result, 4MU treatment decreases tumor growth and increased animal survival. Systemic levels of VEGF were significantly inhibited in 4MU-treated mice. Expression of CD31 was reduced after 4MU therapy in liver parenchyma in comparison with control group. In addition, mRNA expression and protein levels of IL-6 and VEGF were inhibited both in tumor tissue and in nontumoral liver parenchyma. Interestingly, IL-6 production was dramatically reduced in Kupffer cells isolated from 4MU-treated mice, and in Hepa129 cells in vitro. Besides, 4MU was able to inhibit endothelial cell migration and tube formation. In conclusion, 4MU has antitumor activity in vivo and its mechanisms of action involve an inhibition of angiogenesis and IL-6 production. 4MU is an orally available molecule with potential for HCC treatment.
Collapse
Affiliation(s)
- Flavia Piccioni
- Liver Unit, Gene Therapy Laboratory, Facultad de Ciencias Biomédicas, Universidad Austral, Avenida Presidente Perón 1500 (B1629ODT), Derqui-Pilar, Buenos Aires, Argentina
| | - Esteban Fiore
- Liver Unit, Gene Therapy Laboratory, Facultad de Ciencias Biomédicas, Universidad Austral, Avenida Presidente Perón 1500 (B1629ODT), Derqui-Pilar, Buenos Aires, Argentina
| | - Juan Bayo
- Liver Unit, Gene Therapy Laboratory, Facultad de Ciencias Biomédicas, Universidad Austral, Avenida Presidente Perón 1500 (B1629ODT), Derqui-Pilar, Buenos Aires, Argentina
| | - Catalina Atorrasagasti
- Liver Unit, Gene Therapy Laboratory, Facultad de Ciencias Biomédicas, Universidad Austral, Avenida Presidente Perón 1500 (B1629ODT), Derqui-Pilar, Buenos Aires, Argentina CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Buenos Aires, Argentina
| | - Estanislao Peixoto
- Liver Unit, Gene Therapy Laboratory, Facultad de Ciencias Biomédicas, Universidad Austral, Avenida Presidente Perón 1500 (B1629ODT), Derqui-Pilar, Buenos Aires, Argentina
| | - Manglio Rizzo
- Liver Unit, Gene Therapy Laboratory, Facultad de Ciencias Biomédicas, Universidad Austral, Avenida Presidente Perón 1500 (B1629ODT), Derqui-Pilar, Buenos Aires, Argentina
| | - Mariana Malvicini
- Liver Unit, Gene Therapy Laboratory, Facultad de Ciencias Biomédicas, Universidad Austral, Avenida Presidente Perón 1500 (B1629ODT), Derqui-Pilar, Buenos Aires, Argentina CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Buenos Aires, Argentina
| | - Irene Tirado-González
- Laboratory of Reproductive Medicine, Medicine University of Berlin, Charité Centre 12 Internal Medicine and Dermatology, Berlin, Germany
| | - Mariana G García
- Liver Unit, Gene Therapy Laboratory, Facultad de Ciencias Biomédicas, Universidad Austral, Avenida Presidente Perón 1500 (B1629ODT), Derqui-Pilar, Buenos Aires, Argentina CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Buenos Aires, Argentina
| | - Laura Alaniz
- CIT NOBA, Universidad Nacional del Noroeste de la Pcia. de Bs. As., Junín, Buenos Aires, Argentina CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Buenos Aires, Argentina
| | - Guillermo Mazzolini
- Liver Unit, Gene Therapy Laboratory, Facultad de Ciencias Biomédicas, Universidad Austral, Avenida Presidente Perón 1500 (B1629ODT), Derqui-Pilar, Buenos Aires, Argentina CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Buenos Aires, Argentina
| |
Collapse
|
38
|
Kanazawa K, Hagiwara Y, Tsuchiya M, Yabe Y, Sonofuchi K, Koide M, Sekiguchi T, Itaya N, Ando A, Saijo Y, Itoi E. Preventing effects of joint contracture by high molecular weight hyaluronan injections in a rat immobilized knee model. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:3426-3440. [PMID: 26097527 PMCID: PMC4466914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 03/19/2015] [Indexed: 06/04/2023]
Abstract
PURPOSE To elucidate preventive effects of high molecular weight hyaluronan (HMWHA) on the joint capsule of immobilized knees in rats. MATERIALS AND METHODS Unilateral knee joints of rats were immobilized with an internal fixator. Either 50 μl of HMWHA (Im-HA group) or 50 μl of saline (control group) was administered intra-articularly once a week after surgery. Sagittal sections were prepared from the medial midcondylar region of the knee joints and assessed by histological, histomorphometric, and immunohistochemical methods. Gene expressions related to inflammation, fibrotic conditions, and hypoxia were evaluated by quantitative reverse transcription polymerase chain reaction (qRT-PCR). Tissue elasticity of the capsule from both groups was examined using a scanning acoustic microscope (SAM). RESULTS CD68 positive cells decreased in adhesion areas of the synovial membrane after 1 week in both groups. The length of the superficial layer in the synovial membrane of the Im-HA group was significantly longer than those in the control group over a period of 4 to 8 weeks with significantly small numbers of CD68 positive cells. The gene expressions of IL-6, IL-1β, TGF-β, CTGF, COL1a1, COL3a1, SPARC, and HIF1-α were significantly lower in the Im-HA group compared to those in the control group. The sound speed of the anterior and posterior synovial membrane increased significantly (a reduction in elasticity) in the control group compared to those in the Im-HA group during weeks 1 to 4. CONCLUSIONS This study demonstrated that HMWHA injections suppressed inflammatory, fibrotic, and hypoxic conditions observed in the immobilized joint capsule.
Collapse
Affiliation(s)
- Kenji Kanazawa
- Department of Orthopaedic Surgery, Tohoku University School of Medicine1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Yoshihiro Hagiwara
- Department of Orthopaedic Surgery, Tohoku University School of Medicine1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Masahiro Tsuchiya
- Divisions of Aging and Geriatric Dentistry, Tohoku University Graduate School of Dentistry4-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Yutaka Yabe
- Department of Orthopaedic Surgery, Tohoku University School of Medicine1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Kazuaki Sonofuchi
- Department of Orthopaedic Surgery, Tohoku University School of Medicine1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Masashi Koide
- Department of Orthopaedic Surgery, Tohoku University School of Medicine1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Takuya Sekiguchi
- Department of Orthopaedic Surgery, Tohoku University School of Medicine1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Nobuyuki Itaya
- Department of Orthopaedic Surgery, Tohoku University School of Medicine1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Akira Ando
- Department of Orthopaedic Surgery, Tohoku University School of Medicine1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Yoshifumi Saijo
- Department of Biomedical Imaging, Tohoku University Graduate School of Biomedical Engineering4-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Eiji Itoi
- Department of Orthopaedic Surgery, Tohoku University School of Medicine1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| |
Collapse
|
39
|
Peixoto E, Atorrasagasti C, Aquino JB, Militello R, Bayo J, Fiore E, Piccioni F, Salvatierra E, Alaniz L, García MG, Bataller R, Corrales F, Gidekel M, Podhajcer O, Colombo MI, Mazzolini G. SPARC (secreted protein acidic and rich in cysteine) knockdown protects mice from acute liver injury by reducing vascular endothelial cell damage. Gene Ther 2015; 22:9-19. [PMID: 25410742 PMCID: PMC4415270 DOI: 10.1038/gt.2014.102] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Revised: 09/19/2014] [Accepted: 09/24/2014] [Indexed: 12/24/2022]
Abstract
Secreted protein, acidic and rich in cysteine (SPARC) is involved in many biological process including liver fibrogenesis, but its role in acute liver damage is unknown. To examine the role of SPARC in acute liver injury, we used SPARC knock-out (SPARC(-/-)) mice. Two models of acute liver damage were used: concanavalin A (Con A) and the agonistic anti-CD95 antibody Jo2. SPARC expression levels were analyzed in liver samples from patients with acute-on-chronic alcoholic hepatitis (AH). SPARC expression is increased on acute-on-chronic AH patients. Knockdown of SPARC decreased hepatic damage in the two models of liver injury. SPARC(-/-) mice showed a marked reduction in Con A-induced necroinflammation. Infiltration by CD4+ T cells, expression of tumor necrosis factor-α and interleukin-6 and apoptosis were attenuated in SPARC(-/-) mice. Sinusoidal endothelial cell monolayer was preserved and was less activated in Con A-treated SPARC(-/-) mice. SPARC knockdown reduced Con A-induced autophagy of cultured human microvascular endothelial cells (HMEC-1). Hepatic transcriptome analysis revealed several gene networks that may have a role in the attenuated liver damaged found in Con A-treated SPARC(-/-) mice. SPARC has a significant role in the development of Con A-induced severe liver injury. These results suggest that SPARC could represent a therapeutic target in acute liver injury.
Collapse
Affiliation(s)
- E Peixoto
- Liver Unit, Gene Therapy Laboratory, Facultad de Ciencias Biomédicas, Universidad Austral, Buenos Aires, Argentina
| | - C Atorrasagasti
- Liver Unit, Gene Therapy Laboratory, Facultad de Ciencias Biomédicas, Universidad Austral, Buenos Aires, Argentina
- CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Buenos Aires, Argentina
| | - JB Aquino
- Liver Unit, Gene Therapy Laboratory, Facultad de Ciencias Biomédicas, Universidad Austral, Buenos Aires, Argentina
- CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Buenos Aires, Argentina
| | - R Militello
- Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - J Bayo
- Liver Unit, Gene Therapy Laboratory, Facultad de Ciencias Biomédicas, Universidad Austral, Buenos Aires, Argentina
| | - E Fiore
- Liver Unit, Gene Therapy Laboratory, Facultad de Ciencias Biomédicas, Universidad Austral, Buenos Aires, Argentina
| | - F Piccioni
- Liver Unit, Gene Therapy Laboratory, Facultad de Ciencias Biomédicas, Universidad Austral, Buenos Aires, Argentina
| | - E Salvatierra
- Molecular and Cellular Therapy Laboratory, Fundación Instituto Leloir, Buenos Aires, Argentina
| | - L Alaniz
- Liver Unit, Gene Therapy Laboratory, Facultad de Ciencias Biomédicas, Universidad Austral, Buenos Aires, Argentina
- CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Buenos Aires, Argentina
| | - MG García
- Liver Unit, Gene Therapy Laboratory, Facultad de Ciencias Biomédicas, Universidad Austral, Buenos Aires, Argentina
- CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Buenos Aires, Argentina
| | - R Bataller
- University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Institut d’Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - F Corrales
- CIMA, Universidad de Navarra, Pamplona, España
| | - M Gidekel
- Universidad de la Frontera, Temuco, Chile
- Universidad Autónoma de Chile, Santiago, Chile
| | - O Podhajcer
- Molecular and Cellular Therapy Laboratory, Fundación Instituto Leloir, Buenos Aires, Argentina
| | - MI Colombo
- CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Buenos Aires, Argentina
- Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - G Mazzolini
- Liver Unit, Gene Therapy Laboratory, Facultad de Ciencias Biomédicas, Universidad Austral, Buenos Aires, Argentina
- CONICET (Consejo Nacional de Investigaciones Científicas y Técnicas), Buenos Aires, Argentina
| |
Collapse
|
40
|
Huang Y, Liu W, Xiao H, Maitikabili A, Lin Q, Wu T, Huang Z, Liu F, Luo Q, Ouyang G. Matricellular protein periostin contributes to hepatic inflammation and fibrosis. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 185:786-97. [PMID: 25541330 DOI: 10.1016/j.ajpath.2014.11.002] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Revised: 09/29/2014] [Accepted: 11/05/2014] [Indexed: 01/07/2023]
Abstract
Periostin actively contributes to tissue injury, fibrosis, atherosclerosis, and inflammatory diseases; however, its role in hepatic fibrosis is unclear. Herein, we revealed that periostin expression was significantly up-regulated in carbon tetrachloride- and bile duct ligation-induced mice with acute and chronic liver fibrosis. Deficiency in periostin abrogated the development of liver fibrosis in mice. Carbon tetrachloride treatment significantly increased α-smooth muscle actin, fibronectin, and collagen I levels in wild-type mice, which were unaffected in periostin-knockout mice. Periostin-deficient mice showed a significantly reduced area of collagen deposition and decreased levels of serum alanine aminotransferase and aspartate aminotransferase compared with wild-type mice after 2 weeks of carbon tetrachloride administration. Chemokine ligand 2, IL-6, IL-1β, tumor necrosis factor-α, and tissue inhibitor of metalloproteinases 1 mRNA levels were significantly lower in periostin-deficient mice than in wild-type mice after carbon tetrachloride treatment. Periostin colocalized with hepatic stellate cell-derived collagen I and α-smooth muscle actin in mouse acute and chronic fibrotic liver tissues. Transforming growth factor (TGF)-β1 markedly induced periostin expression in primary mouse hepatic stellate cells. Periostin-deficient mice showed significantly lower levels of TGF-β1 and TGF-β2 compared with wild-type mice after carbon tetrachloride treatment. High levels of periostin in patients with acute or chronic hepatitis correlated with TGF-β1 and TGF-β2 expression in serum from patients with hepatitis. Data indicate that periostin is a novel mediator of hepatic fibrosis development.
Collapse
Affiliation(s)
- Yangmei Huang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, China
| | - Weiping Liu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, China
| | - Hongjun Xiao
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, China
| | - Alaiyi Maitikabili
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, China
| | - Qinghua Lin
- Clinical Laboratory, Xiamen Hospital of Traditional Chinese Medicine, Xiamen, China
| | - Tiantian Wu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, China
| | - Zhengjie Huang
- Department of Surgical Oncology, First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Fan Liu
- Medical College, Xiamen University, Xiamen, China
| | - Qi Luo
- Department of Surgical Oncology, First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Gaoliang Ouyang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, China.
| |
Collapse
|
41
|
Rybinski B, Franco-Barraza J, Cukierman E. The wound healing, chronic fibrosis, and cancer progression triad. Physiol Genomics 2014; 46:223-44. [PMID: 24520152 PMCID: PMC4035661 DOI: 10.1152/physiolgenomics.00158.2013] [Citation(s) in RCA: 172] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 02/04/2014] [Indexed: 02/07/2023] Open
Abstract
For decades tumors have been recognized as "wounds that do not heal." Besides the commonalities that tumors and wounded tissues share, the process of wound healing also portrays similar characteristics with chronic fibrosis. In this review, we suggest a tight interrelationship, which is governed as a concurrence of cellular and microenvironmental reactivity among wound healing, chronic fibrosis, and cancer development/progression (i.e., the WHFC triad). It is clear that the same cell types, as well as soluble and matrix elements that drive wound healing (including regeneration) via distinct signaling pathways, also fuel chronic fibrosis and tumor progression. Hence, here we review the relationship between fibrosis and cancer through the lens of wound healing.
Collapse
Affiliation(s)
- Brad Rybinski
- Cancer Biology Program, Fox Chase Cancer Center/Temple Health, Philadelphia, Pennsylvania
| | | | | |
Collapse
|
42
|
Noto Llana M, Sarnacki SH, Aya Castañeda MDR, Bernal MI, Giacomodonato MN, Cerquetti MC. Consumption of Lactobacillus casei fermented milk prevents Salmonella reactive arthritis by modulating IL-23/IL-17 expression. PLoS One 2013; 8:e82588. [PMID: 24340048 PMCID: PMC3858332 DOI: 10.1371/journal.pone.0082588] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Accepted: 10/25/2013] [Indexed: 12/11/2022] Open
Abstract
Reactive arthritis is the development of sterile joint inflammation as a sequel to a remote infection, often in the gut. We have previously shown that a low dose of S. enteritidis inoculated to streptomycin-pretreated mice generates a self-limiting enterocolitis suitable for studying reactive arthritis. Here we show that consumption of Lactobacillus casei prior to infection abolishes intestinal and joint inflammation triggered by Salmonella. BALB/c mice were sacrificed after infection; intestinal and joint samples were analyzed for histological changes and expression of cytokines. TNF-α was measured by ELISA and the expression of IL-1β, IL-6, IL-10, IL-17, IL-23 and TGF-β was assessed by qPCR. L. casei consumption prevented Salmonella-induced synovitis, the increment of TNF-α in knees and the increase of IL-17 expression in popliteal and inguinal lymph nodes. At intestinal level consumption of L. casei drastically diminished S. enteritidis invasiveness and shortened splenic persistence of the pathogen. Bacterial loads recovered at days 2 and 5 from Peyer's patches were 10-fold lower in mice fed with L. casei. In accordance, we found that the augment in gut permeability induced during enterocolitis was decreased in those animals. Consumption of L. casei prior to infection failed to increase anti- inflammatory molecules such as IL-10 and TGF-β in the intestine. On the other hand, consumption of L. casei abrogated the expression of TNF-α, IL-17, IL-23, IL-1β and IL-6 in cecum and mesenteric lymph nodes. These cytokines are needed for differentiation of immune cells involved in the development of reactive arthritis such as Th17 and γδ T cells. Trafficking of these inflammatory cells from the gut to the joints has been proposed as a mechanism of generation of reactive arthritis. Our results suggest that L. casei consumption prevents Salmonella-induced synovitis by altering the intestinal milieu necessary for differentiation of cells involved in the generation of joint inflammation.
Collapse
Affiliation(s)
- Mariángeles Noto Llana
- Instituto de Microbiología y Parasitología Médica (IMPaM-CONICET) and Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Sebastián Hernán Sarnacki
- Instituto de Microbiología y Parasitología Médica (IMPaM-CONICET) and Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María del Rosario Aya Castañeda
- Instituto de Microbiología y Parasitología Médica (IMPaM-CONICET) and Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María Isabel Bernal
- Instituto de Microbiología y Parasitología Médica (IMPaM-CONICET) and Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Mónica Nancy Giacomodonato
- Instituto de Microbiología y Parasitología Médica (IMPaM-CONICET) and Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María Cristina Cerquetti
- Instituto de Microbiología y Parasitología Médica (IMPaM-CONICET) and Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
43
|
Downregulation of SPARC expression inhibits the invasion of human trophoblast cells in vitro. PLoS One 2013; 8:e69079. [PMID: 23935929 PMCID: PMC3720866 DOI: 10.1371/journal.pone.0069079] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Accepted: 06/04/2013] [Indexed: 01/10/2023] Open
Abstract
Successful pregnancy depends on the precise regulation of extravilloustrophoblast (EVT) invasion into the uterine decidua. SPARC (secreted protein acidic and rich in cysteine) is a matricellular glycoprotein that plays critical roles in the pathologies associated with obesity and diabetes, as well as tumorigenesis. The objective of this study was to investigate the role of SPARC in the process of trophoblast invasion which shares many similarities with tumor cell invasion. By Western blot, higher expression of SPARC was observed in mouse brain, ovary and uterus compared to other mouse tissues. Immunohistochemistry analysis revealed a spatio-temporal expression of SPARC in mouse uterus in the periimplantation period. At the implantation site of d8 pregnancy, SPARC mainly accumulated in the secondary decidua zone (SDZ), trophoblast cells and blastocyst. The expression of SPARC was also detected in human placental villi and trophoblast cell lines. In a Matrigel invasion assay, we found SPARC-specific RNA interference significantly reduced the invasion of human extravilloustrophoblast HTR8/SVneo cells. Microarray analysis revealed that SPARC depletion upregulated the expression of interleukin 11 (IL11), KISS1, insulin-like growth factor binding protein 4 (IGFBP4), collagen type I alpha 1 (COLIA1), matrix metallopeptidase 9 (MMP9), and downregulated the expression of the alpha polypeptide of chorionic gonadotropin (CGA), MMP1, gap junction protein alpha 1 (GJA1), et al. The gene array result was further validated by qRT-PCR and Western blot. The present data indicate that SPARC may play an important role in the regulation of normal placentation by promoting the invasion of trophoblast cells into the uterine decidua.
Collapse
|