1
|
Baquero C, Iniesta‐González M, Palao N, Fernández‐Infante C, Cueto‐Remacha M, Mancebo J, de la Cámara‐Fuentes S, Rodrigo‐Faus M, Valdecantos MP, Valverde AM, Sequera C, Manzano S, Cuesta ÁM, Gutierrez‐Uzquiza A, Bragado P, Guerrero C, Porras A. Platelet C3G protects from liver fibrosis, while enhancing tumor growth through regulation of the immune response. J Pathol 2025; 265:502-517. [PMID: 39989399 PMCID: PMC11880977 DOI: 10.1002/path.6403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/29/2024] [Accepted: 01/09/2025] [Indexed: 02/25/2025]
Abstract
Primary liver cancer usually occurs in the context of chronic liver disease (CLD), in association with fibrosis. Platelets have emerged as important regulators of CLD and liver cancer, although their precise function and mechanism of action need to be clarified. C3G (RapGEF1) regulates platelet activation, adhesion, and secretion. Here we evaluate the role of platelet C3G in chemically induced fibrosis and liver cancer associated with fibrosis using genetically modified mouse models. We found that while overexpression of full-length C3G in platelets decreased liver fibrosis induced by chronic treatment with CCl4, overexpressed C3G lacking the catalytic domain did not, although in both cases platelet recruitment to the liver was similar. In addition, C3G deletion in platelets (PF4-C3GKO mouse model) increased CCl4-induced liver damage and hepatic fibrosis, reducing liver platelets and macrophages. Moreover, early liver immune response to CCl4 was altered in PF4-C3GKO mice, with a remarkable lower activation of macrophages and increased monocyte-derived macrophages compared to WT mice. On the other hand, in response to DEN+CCl4, PF4-C3G WT mice exhibited more and larger liver tumors than PF4-C3GKO mice, accompanied by the presence of more platelets, despite having less fibrosis in previous steps. Liver immune cell populations were also differentially regulated in PF4-C3GKO mice, highlighting the higher number of macrophages, likely with a pro-inflammatory phenotype, present in the liver in response to chronic DEN+CCl4 treatment. Proteins upregulated or downregulated in platelet-rich plasma from PF4-C3GKO compared to WT mice might regulate the immune response and tumor development. In this regard, enrichment analyses using proteomic data showed changes in several proteins involved in platelet activation and immune response pathways. Additionally, the higher secretion of CD40L by PF4-C3GKO platelets could contribute to their antitumor effect. Therefore, platelet C3G presents antifibrotic and protumor effects in the liver, likely mediated by changes in the immune response. © 2025 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Cristina Baquero
- Departamento de Bioquímica y Biología Molecular, Facultad de FarmaciaUniversidad Complutense de MadridMadridSpain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC)MadridSpain
| | - Minerva Iniesta‐González
- Departamento de Bioquímica y Biología Molecular, Facultad de FarmaciaUniversidad Complutense de MadridMadridSpain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC)MadridSpain
| | - Nerea Palao
- Departamento de Bioquímica y Biología Molecular, Facultad de FarmaciaUniversidad Complutense de MadridMadridSpain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC)MadridSpain
| | - Cristina Fernández‐Infante
- Instituto de Biología Molecular y Celular del Cáncer (IMBCC)Universidad de Salamanca‐CSICSalamancaSpain
- Instituto de Investigación Biomédica de Salamanca (IBSAL)SalamancaSpain
| | - Mateo Cueto‐Remacha
- Departamento de Bioquímica y Biología Molecular, Facultad de FarmaciaUniversidad Complutense de MadridMadridSpain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC)MadridSpain
| | - Jaime Mancebo
- Departamento de Bioquímica y Biología Molecular, Facultad de FarmaciaUniversidad Complutense de MadridMadridSpain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC)MadridSpain
| | | | - María Rodrigo‐Faus
- Departamento de Bioquímica y Biología Molecular, Facultad de FarmaciaUniversidad Complutense de MadridMadridSpain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC)MadridSpain
| | - M Pilar Valdecantos
- Instituto de Investigaciones Biomédicas (IIBM) Alberto Sols‐Morreale (CSIC‐UAM)MadridSpain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERdem)Instituto de Salud Carlos IIIMadridSpain
| | - Angela M Valverde
- Instituto de Investigaciones Biomédicas (IIBM) Alberto Sols‐Morreale (CSIC‐UAM)MadridSpain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERdem)Instituto de Salud Carlos IIIMadridSpain
| | - Celia Sequera
- Departamento de Bioquímica y Biología Molecular, Facultad de FarmaciaUniversidad Complutense de MadridMadridSpain
- Aix Marseille Univ, CNRS, InsermInstitut Paoli‐Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM)MarseilleFrance
| | - Sara Manzano
- Departamento de Bioquímica y Biología Molecular, Facultad de FarmaciaUniversidad Complutense de MadridMadridSpain
| | - Ángel M Cuesta
- Departamento de Bioquímica y Biología Molecular, Facultad de FarmaciaUniversidad Complutense de MadridMadridSpain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC)MadridSpain
| | - Alvaro Gutierrez‐Uzquiza
- Departamento de Bioquímica y Biología Molecular, Facultad de FarmaciaUniversidad Complutense de MadridMadridSpain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC)MadridSpain
| | - Paloma Bragado
- Departamento de Bioquímica y Biología Molecular, Facultad de FarmaciaUniversidad Complutense de MadridMadridSpain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC)MadridSpain
| | - Carmen Guerrero
- Instituto de Biología Molecular y Celular del Cáncer (IMBCC)Universidad de Salamanca‐CSICSalamancaSpain
- Instituto de Investigación Biomédica de Salamanca (IBSAL)SalamancaSpain
- Departamento de MedicinaUniversidad de SalamancaSalamancaSpain
| | - Almudena Porras
- Departamento de Bioquímica y Biología Molecular, Facultad de FarmaciaUniversidad Complutense de MadridMadridSpain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC)MadridSpain
| |
Collapse
|
2
|
Sererols-Viñas L, Garcia-Vicién G, Ruiz-Blázquez P, Lee TF, Lee YA, Gonzalez-Sanchez E, Vaquero J, Moles A, Filliol A, Affò S. Hepatic Stellate Cells Functional Heterogeneity in Liver Cancer. Semin Liver Dis 2025; 45:33-51. [PMID: 40043738 DOI: 10.1055/a-2551-0724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/01/2025]
Abstract
Hepatic stellate cells (HSCs) are the liver's pericytes, and play key roles in liver homeostasis, regeneration, fibrosis, and cancer. Upon injury, HSCs activate and are the main origin of myofibroblasts and cancer-associated fibroblasts (CAFs) in liver fibrosis and cancer. Primary liver cancer has a grim prognosis, ranking as the third leading cause of cancer-related deaths worldwide, with hepatocellular carcinoma (HCC) being the predominant type, followed by intrahepatic cholangiocarcinoma (iCCA). Moreover, the liver hosts 35% of all metastatic lesions. The distinct spatial distribution and functional roles of HSCs across these malignancies represent a significant challenge for universal therapeutic strategies, requiring a nuanced and tailored understanding of their contributions. This review examines the heterogeneous roles of HSCs in liver cancer, focusing on their spatial localization, dynamic interactions within the tumor microenvironment (TME), and emerging therapeutic opportunities, including strategies to modulate their activity, and harness their potential as targets for antifibrotic and antitumor interventions.
Collapse
Affiliation(s)
- Laura Sererols-Viñas
- Tumor Microenvironment Plasticity and Heterogeneity Research Group, Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- University of Barcelona, Barcelona, Spain
| | - Gemma Garcia-Vicién
- Tumor Microenvironment Plasticity and Heterogeneity Research Group, Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Paloma Ruiz-Blázquez
- University of Barcelona, Barcelona, Spain
- Tissue Remodeling Fibrosis and Cancer Group, Institute of Biomedical Research of Barcelona, Spanish National Research Council, Barcelona, Spain
- Institute of Biomedical Research of Barcelona (IDIBAPS), Barcelona, Spain
- CIBEREHD, National Biomedical Research Institute on Liver and Gastrointestinal Diseases, Instituto de Salud Carlos III, Madrid, Spain
| | - Ting-Fang Lee
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Youngmin A Lee
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Ester Gonzalez-Sanchez
- HepatoBiliary Tumours Lab, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, CSIC-Universidad de Salamanca, Salamanca, Spain
- Department of Physiology and Pharmacology, University of Salamanca, Salamanca, Spain
| | - Javier Vaquero
- CIBEREHD, National Biomedical Research Institute on Liver and Gastrointestinal Diseases, Instituto de Salud Carlos III, Madrid, Spain
- HepatoBiliary Tumours Lab, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, CSIC-Universidad de Salamanca, Salamanca, Spain
- TGF-β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - Anna Moles
- Tissue Remodeling Fibrosis and Cancer Group, Institute of Biomedical Research of Barcelona, Spanish National Research Council, Barcelona, Spain
- Institute of Biomedical Research of Barcelona (IDIBAPS), Barcelona, Spain
- CIBEREHD, National Biomedical Research Institute on Liver and Gastrointestinal Diseases, Instituto de Salud Carlos III, Madrid, Spain
| | - Aveline Filliol
- Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Silvia Affò
- Tumor Microenvironment Plasticity and Heterogeneity Research Group, Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| |
Collapse
|
3
|
Nouri K, Piryaei A, Seydi H, Zarkesh I, Ghoytasi I, Shokouhian B, Najimi M, Vosough M. Fibrotic liver extracellular matrix induces cancerous phenotype in biomimetic micro-tissues of hepatocellular carcinoma model. Hepatobiliary Pancreat Dis Int 2025; 24:92-103. [PMID: 39289044 DOI: 10.1016/j.hbpd.2024.09.003] [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: 02/06/2024] [Accepted: 07/25/2024] [Indexed: 09/19/2024]
Abstract
BACKGROUND Despite considerable advancements in identifying factors contributing to the development of hepatocellular carcinoma (HCC), the pathogenesis of HCC remains unclear. In many cases, HCC is a consequence of prolonged liver fibrosis, resulting in the formation of an intricate premalignant microenvironment. The accumulation of extracellular matrix (ECM) is a hallmark of premalignant microenvironment. Given the critical role of different matrix components in regulating cell phenotype and function, this study aimed to elucidate the interplay between the fibrotic matrix and malignant features in HCC. METHODS Liver tissues from both control (normal) and carbon tetrachloride (CCl4)-induced fibrotic rats were decellularized using sodium dodecyl sulfate (SDS) and Triton X-100. The resulting hydrogel from decellularized ECM was processed into micro-particles via the water-in-oil emulsion method. Micro-particles were subsequently incorporated into three-dimensional liver biomimetic micro-tissues (MTs) comprising Huh-7 cells, human umbilical vein endothelial cells (HUVECs), and LX-2 cells. The MTs were evaluated using the 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) assay at day 11, immunofluorescence staining, immunoblotting, and spheroid migration assay at day 14 after co-culture. RESULTS Fibrotic matrix from CCl4-treated rat livers significantly enhanced the growth rate of the MTs and their expression of CCND1 as compared to the normal one. Fibrotic matrix, also induced the expression of epithelial-to-mesenchymal transition (EMT)-associated genes such as TWIST1, ACTA2, MMP9, CDH2, and VIMENTIN in the MTs as compared to the normal matrix. Conversely, the expression of CDH1 and hepatic maturation genes HNF4A, ALB, CYP3A4 was decreased in the MTs when the fibrotic matrix was used. Furthermore, the fibrotic matrix increased the migration of the MTs and their secretion of alpha-fetoprotein. CONCLUSIONS Our findings suggest a regulatory role for the fibrotic matrix in promoting cancerous phenotype, which could potentially accelerate the progression of malignancy in the liver.
Collapse
Affiliation(s)
- Kosar Nouri
- Department of Developmental Biology, University of Science and Culture, ACECR 14155-4364 Tehran, Iran; Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR 14155-4364 Tehran, Iran; Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR 14155-4364 Tehran, Iran
| | - Abbas Piryaei
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Homeyra Seydi
- Department of Developmental Biology, University of Science and Culture, ACECR 14155-4364 Tehran, Iran; Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR 14155-4364 Tehran, Iran; Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR 14155-4364 Tehran, Iran
| | - Ibrahim Zarkesh
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR 14155-4364 Tehran, Iran
| | - Ibrahim Ghoytasi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR 14155-4364 Tehran, Iran
| | - Bahare Shokouhian
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR 14155-4364 Tehran, Iran; Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mustapha Najimi
- Laboratory of Pediatric Hepatology and Cell Therapy, Institute of Experimental and Clinical Research (IREC), UCLouvain, Brussels, Belgium
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR 14155-4364 Tehran, Iran; Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
4
|
Hu H, Yin Y, Zhou H, Jiang B, Cai T, Wu S, Guo S. Umbilical cord mesenchymal stem cell-derived exosomal Follistatin inhibits fibrosis and promotes muscle regeneration in mice by influencing Smad2 and AKT signaling. Exp Cell Res 2025; 444:114396. [PMID: 39732451 DOI: 10.1016/j.yexcr.2024.114396] [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: 04/24/2024] [Revised: 12/22/2024] [Accepted: 12/25/2024] [Indexed: 12/30/2024]
Abstract
BACKGROUND Promoting muscle regeneration through stem cell therapy has potential risks. We investigated the effect of umbilical cord mesenchymal stem cells (UMSCs) Exosomes (Exo) Follistatin on muscle regeneration. METHODS The Exo was derived from UMSCs cells and was utilized to affect the mice muscle injury model and C2C12 cells myotubes atrophy model. The Western blot, qRT-PCR and IF were utilized to determine the effects of Exo on the levels of Follistatin, MyHC, MyoD, Myostatin, MuRF1, MAFbx, α-SMA, Collagen I, Smad2, and AKT. In addition, HE and Masson staining were used to assess muscle tissue damage in mice. RESULTS The level of Follistatin in Exo was significantly higher than that in UMSCs. UMSCs-Exo increased the levels of Follistatin, MyHC, MyoD, and p-Smad2 and decreased the levels of Myostatin, MuRF1, MAFbx, α-SMA, Collagen I, p-AKT, and p-mTOR in mice or C2C12 cells. In addition, UMSCs-Exo decreased levels of inflammation and fibrosis in mice. However, UMSCs-Exo-si-Follistatin reversed the effect of UMSCs-Exo. Transfection of oe-Smad2 up-regulated the protein levels of Collagen I, α-SMA, and changed the ratio of p-Smad2/Smad2 expression to 0.33, and 0.34, 0.73. LY294002 decreased the levels of MyHC, MyoD, and the ratio of p-AKT/AKT and p-mTOR/mTOR expression to 0.12, 0.17, 0.33, and 0.41, increased the levels of MuRF1 and MAFbx to 0.36 and 0.34. CONCLUSION This study demonstrated that Follistatin in UMSCs-Exo inhibits fibrosis and promotes muscle regeneration in mice by regulating Smad and AKT signaling.
Collapse
Affiliation(s)
- Hai Hu
- Department of Orthopedics, The Third Xiangya Hospital, Central South University, Changsha, China; Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research on Functional Nucleic Acid, Changsha Medical University, Changsha, China
| | - Yuesong Yin
- Department of Orthopedics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Hecheng Zhou
- Department of Orthopedics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Binbin Jiang
- Department of Orthopedics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Ting Cai
- Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research on Functional Nucleic Acid, Changsha Medical University, Changsha, China
| | - Song Wu
- Department of Orthopedics, The Third Xiangya Hospital, Central South University, Changsha, China.
| | - Shuangfei Guo
- Department of Limbs (Foot and Hand) Microsurgery, Chenzhou No.1 People's Hospital, The First Clinical Medical College Affiliated to Southern Medical University, Chenzhou, Hunan, China.
| |
Collapse
|
5
|
Chen Z, Long L, Wang J, Jiang M, Li W, Cui W, Zou L. Enhanced Tumor Site Accumulation and Therapeutic Efficacy of Extracellular Matrix-Drug Conjugates Targeting Tumor Cells. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2402040. [PMID: 38829027 DOI: 10.1002/smll.202402040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/14/2024] [Indexed: 06/05/2024]
Abstract
The extracellular matrix (ECM) engages in regulatory interactions with cell surface receptors through its constituent proteins and polysaccharides. Therefore, nano-sized extracellular matrix conjugated with doxorubicin (DOX) is utilized to produce extracellular matrix-drug conjugates (ECM-DOX) tailored for targeted delivery to cancer cells. The ECM-DOX nanoparticles exhibit rod-like morphology, boasting a commendable drug loading capacity of 4.58%, coupled with acid-sensitive drug release characteristics. Notably, ECM-DOX nanoparticles enhance the uptake by tumor cells and possess the ability to penetrate endothelial cells and infiltrate tumor multicellular spheroids. Mechanistic insights reveal that the internalization of ECM-DOX nanoparticle is facilitated through clathrin-mediated endocytosis and macropinocytosis, intricately involving hyaluronic acid receptors and integrins. Pharmacokinetic assessments unveil a prolonged blood half-life of ECM-DOX nanoparticles at 3.65 h, a substantial improvement over the 1.09 h observed for free DOX. A sustained accumulation effect of ECM-DOX nanoparticles at tumor sites, with drug levels in tumor tissues surpassing those of free DOX by several-fold. The profound therapeutic impact of ECM-DOX nanoparticles is evident in their notable inhibition of tumor growth, extension of median survival time in animals, and significant reduction in DOX-induced cardiotoxicity. The ECM platform emerges as a promising carrier for avant-garde nanomedicines in the realm of cancer treatment.
Collapse
Affiliation(s)
- Zhoujiang Chen
- Institute for Advanced Study, Chengdu University, Chengdu, Sichuan, 610106, P. R China
| | - Lianlin Long
- School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, 563099, P. R China
| | - Ji Wang
- School of Pharmacy, Chengdu University, Chengdu, Sichuan, 610106, P. R. China
| | - Mingli Jiang
- School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, 563099, P. R China
| | - Wei Li
- School of Pharmacy, Chengdu University, Chengdu, Sichuan, 610106, P. R. China
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R China
| | - Liang Zou
- Institute for Advanced Study, Chengdu University, Chengdu, Sichuan, 610106, P. R China
| |
Collapse
|
6
|
Macdonald JK, Taylor HB, Wang M, Delacourt A, Edge C, Lewin DN, Kubota N, Fujiwara N, Rasha F, Marquez CA, Ono A, Oka S, Chayama K, Lewis S, Taouli B, Schwartz M, Fiel MI, Drake RR, Hoshida Y, Mehta AS, Angel PM. The Spatial Extracellular Proteomic Tumor Microenvironment Distinguishes Molecular Subtypes of Hepatocellular Carcinoma. J Proteome Res 2024; 23:3791-3805. [PMID: 38980715 PMCID: PMC11385377 DOI: 10.1021/acs.jproteome.4c00099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 05/31/2024] [Accepted: 06/15/2024] [Indexed: 07/10/2024]
Abstract
Hepatocellular carcinoma (HCC) mortality rates continue to increase faster than those of other cancer types due to high heterogeneity, which limits diagnosis and treatment. Pathological and molecular subtyping have identified that HCC tumors with poor outcomes are characterized by intratumoral collagenous accumulation. However, the translational and post-translational regulation of tumor collagen, which is critical to the outcome, remains largely unknown. Here, we investigate the spatial extracellular proteome to understand the differences associated with HCC tumors defined by Hoshida transcriptomic subtypes of poor outcome (Subtype 1; S1; n = 12) and better outcome (Subtype 3; S3; n = 24) that show differential stroma-regulated pathways. Collagen-targeted mass spectrometry imaging (MSI) with the same-tissue reference libraries, built from untargeted and targeted LC-MS/MS was used to spatially define the extracellular microenvironment from clinically-characterized, formalin-fixed, paraffin-embedded tissue sections. Collagen α-1(I) chain domains for discoidin-domain receptor and integrin binding showed distinctive spatial distribution within the tumor microenvironment. Hydroxylated proline (HYP)-containing peptides from the triple helical regions of fibrillar collagens distinguished S1 from S3 tumors. Exploratory machine learning on multiple peptides extracted from the tumor regions could distinguish S1 and S3 tumors (with an area under the receiver operating curve of ≥0.98; 95% confidence intervals between 0.976 and 1.00; and accuracies above 94%). An overall finding was that the extracellular microenvironment has a high potential to predict clinically relevant outcomes in HCC.
Collapse
Affiliation(s)
- Jade K. Macdonald
- Department
of Cell and Molecular Pharmacology, Medical
University of South Carolina, Charleston, South Carolina 29425, United States
| | - Harrison B. Taylor
- Department
of Cell and Molecular Pharmacology, Medical
University of South Carolina, Charleston, South Carolina 29425, United States
| | - Mengjun Wang
- Department
of Cell and Molecular Pharmacology, Medical
University of South Carolina, Charleston, South Carolina 29425, United States
| | - Andrew Delacourt
- Department
of Cell and Molecular Pharmacology, Medical
University of South Carolina, Charleston, South Carolina 29425, United States
| | - Christin Edge
- Department
of Cell and Molecular Pharmacology, Medical
University of South Carolina, Charleston, South Carolina 29425, United States
| | - David N. Lewin
- Department
of Cell and Molecular Pharmacology, Medical
University of South Carolina, Charleston, South Carolina 29425, United States
| | - Naoto Kubota
- Liver
Tumor Translational Research Program, Simmons Comprehensive Cancer
Center, Division of Digestive and Liver Diseases, Department of Internal
Medicine, University of Texas Southwestern
Medical Center, Dallas, Texas 75390, United States
| | - Naoto Fujiwara
- Liver
Tumor Translational Research Program, Simmons Comprehensive Cancer
Center, Division of Digestive and Liver Diseases, Department of Internal
Medicine, University of Texas Southwestern
Medical Center, Dallas, Texas 75390, United States
| | - Fahmida Rasha
- Liver
Tumor Translational Research Program, Simmons Comprehensive Cancer
Center, Division of Digestive and Liver Diseases, Department of Internal
Medicine, University of Texas Southwestern
Medical Center, Dallas, Texas 75390, United States
| | - Cesia A. Marquez
- Liver
Tumor Translational Research Program, Simmons Comprehensive Cancer
Center, Division of Digestive and Liver Diseases, Department of Internal
Medicine, University of Texas Southwestern
Medical Center, Dallas, Texas 75390, United States
| | - Atsushi Ono
- Department
of Gastroenterology, Graduate School of
Biomedical & Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Shiro Oka
- Department
of Gastroenterology, Graduate School of
Biomedical & Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Kazuaki Chayama
- Hiroshima
Institute of Life Sciences, Hiroshima 734-8553, Japan
- Collaborative
Research Laboratory of Medical Innovation, Research Center for Hepatology
and Gastroenterology, Hiroshima University, Hiroshima 734-8553, Japan
- RIKEN Center
for Integrative Medical Sciences, Yokohama 230-0045, Japan
| | - Sara Lewis
- Department
of Radiology, Icahn School of Medicine at
Mount Sinai, New York, New York 10029, United States
| | - Bachir Taouli
- Department
of Radiology, Icahn School of Medicine at
Mount Sinai, New York, New York 10029, United States
| | - Myron Schwartz
- Department
of Radiology, Icahn School of Medicine at
Mount Sinai, New York, New York 10029, United States
- Department
of Surgery, Icahn School of Medicine at
Mount Sinai, New York, New York 10029, United States
| | - M Isabel Fiel
- Department
of Radiology, Icahn School of Medicine at
Mount Sinai, New York, New York 10029, United States
- Department
of Pathology, Icahn School of Medicine at
Mount Sinai, New York, New York 10029, United States
| | - Richard R. Drake
- Department
of Cell and Molecular Pharmacology, Medical
University of South Carolina, Charleston, South Carolina 29425, United States
| | - Yujin Hoshida
- Liver
Tumor Translational Research Program, Simmons Comprehensive Cancer
Center, Division of Digestive and Liver Diseases, Department of Internal
Medicine, University of Texas Southwestern
Medical Center, Dallas, Texas 75390, United States
| | - Anand S. Mehta
- Department
of Cell and Molecular Pharmacology, Medical
University of South Carolina, Charleston, South Carolina 29425, United States
| | - Peggi M. Angel
- Department
of Cell and Molecular Pharmacology, Medical
University of South Carolina, Charleston, South Carolina 29425, United States
| |
Collapse
|
7
|
Liu J, Liu J, Mu W, Ma Q, Zhai X, Jin B, Liu Y, Zhang N. Delivery Strategy to Enhance the Therapeutic Efficacy of Liver Fibrosis via Nanoparticle Drug Delivery Systems. ACS NANO 2024; 18:20861-20885. [PMID: 39082637 DOI: 10.1021/acsnano.4c02380] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Liver fibrosis (LF) is a pathological repair reaction caused by a chronic liver injury that affects the health of millions of people worldwide, progressing to life-threatening cirrhosis and liver cancer without timely intervention. Due to the complexity of LF pathology, multiple etiological characteristics, and the deposited extracellular matrix, traditional drugs cannot reach appropriate targets in a time-space matching way, thus decreasing the therapeutic effect. Nanoparticle drug delivery systems (NDDS) enable multidrug co-therapy and develop multifactor delivery strategies targeting pathological processes, showing great potential in LF therapy. Based on the pathogenesis and the current clinical treatment status of LF, we systematically elucidate the targeting mechanism of NDDS used in the treatment of LF. Subsequently, we focus on the progress of drug delivery applications for LF, including combined delivery for the liver fibrotic pathological environment, overcoming biological barriers, precise intracellular regulation, and intelligent responsive delivery for the liver fibrotic microenvironment. We hope that this review will inspire the rational design of NDDS for LF in the future in order to provide ideas and methods for promoting LF regression and cure.
Collapse
Affiliation(s)
- Jie Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Jinhu Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Weiwei Mu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Qingping Ma
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Xiangyu Zhai
- Department of Hepatobiliary Surgery, The Second Hospital of Shandong University, Jinan, Shandong 250033, China
| | - Bin Jin
- Department of Hepatobiliary Surgery, The Second Hospital of Shandong University, Jinan, Shandong 250033, China
- Organ Transplant Department, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Yongjun Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Na Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| |
Collapse
|
8
|
Bogomolova A, Balakrishnan A, Ott M, Sharma AD. "The Good, the Bad, and the Ugly" - About Diverse Phenotypes of Hepatic Stellate Cells in the Liver. Cell Mol Gastroenterol Hepatol 2024; 17:607-622. [PMID: 38216053 PMCID: PMC10900761 DOI: 10.1016/j.jcmgh.2024.01.002] [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: 08/07/2023] [Revised: 01/04/2024] [Accepted: 01/04/2024] [Indexed: 01/14/2024]
Abstract
Hepatic stellate cells (HSCs) and their activated derivatives, often referred to as myofibroblasts (MFs), play a key role in progression of chronic liver injuries leading to fibrosis, cirrhosis, and hepatocellular carcinoma. Until recently, MFs were considered a homogenous cell type majorly due to lack of techniques that allow complex molecular studies at a single-cell resolution. Recent technical advancements in genetic lineage-tracing models as well as the exponential growth of studies with single-cell transcriptome and proteome analyses have uncovered hidden heterogeneities among the HSC and MF populations in healthy states as well as chronic liver injuries at the various stages of tissue deformation. The identification of different phenotypes along the HSC/MF axis, which either maintain essential liver functions ("good" HSCs), emerge during fibrosis ("bad" HSCs), or even promote hepatocellular carcinoma ("ugly" HSCs), may lay the foundation for targeting a particular MF phenotype as potential treatment for chronic liver injuries.
Collapse
Affiliation(s)
- Alexandra Bogomolova
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany; Research Group RNA Therapeutics & Liver Regeneration, REBIRTH-Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Asha Balakrishnan
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Michael Ott
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany.
| | - Amar Deep Sharma
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany; Research Group RNA Therapeutics & Liver Regeneration, REBIRTH-Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
9
|
Sabir U, Gu HM, Zhang DW. Extracellular matrix turnover: phytochemicals target and modulate the dual role of matrix metalloproteinases (MMPs) in liver fibrosis. Phytother Res 2023; 37:4932-4962. [PMID: 37461256 DOI: 10.1002/ptr.7959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 06/05/2023] [Accepted: 07/02/2023] [Indexed: 11/10/2023]
Abstract
Extracellular matrix (ECM) resolution by matrix metalloproteinases (MMPs) is a well-documented mechanism. MMPs play a dual and complex role in modulating ECM degradation at different stages of liver fibrosis, depending on the timing and levels of their expression. Increased MMP-1 combats disease progression by cleaving the fibrillar ECM. Activated hepatic stellate cells (HSCs) increase expression of MMP-2, -9, and -13 in different chemicals-induced animal models, which may alleviate or worsen disease progression based on animal models and the stage of liver fibrosis. In the early stage, elevated expression of certain MMPs may damage surrounding tissue and activate HSCs, promoting fibrosis progression. At the later stage, downregulation of MMPs can facilitate ECM accumulation and disease progression. A number of phytochemicals modulate MMP activity and ECM turnover, alleviating disease progression. However, the effects of phytochemicals on the expression of different MMPs are variable and may depend on the disease models and stage, and the dosage, timing and duration of phytochemicals used in each study. Here, we review the most recent advances in the role of MMPs in the effects of phytochemicals on liver fibrogenesis, which indicates that further studies are warranted to confirm and define the potential clinical efficacy of these phytochemicals.
Collapse
Affiliation(s)
- Usman Sabir
- Department of Pediatrics and Group on the Molecular and Cell Biology of Lipids, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Hong-Mei Gu
- Department of Pediatrics and Group on the Molecular and Cell Biology of Lipids, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Da-Wei Zhang
- Department of Pediatrics and Group on the Molecular and Cell Biology of Lipids, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
10
|
Affo S, Filliol A, Gores GJ, Schwabe RF. Fibroblasts in liver cancer: functions and therapeutic translation. Lancet Gastroenterol Hepatol 2023; 8:748-759. [PMID: 37385282 PMCID: PMC11809766 DOI: 10.1016/s2468-1253(23)00111-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/13/2023] [Accepted: 04/14/2023] [Indexed: 07/01/2023]
Abstract
Accumulation of fibroblasts in the premalignant or malignant liver is a characteristic feature of liver cancer, but has not been therapeutically leveraged despite evidence for pathophysiologically relevant roles in tumour growth. Hepatocellular carcinoma is a largely non-desmoplastic tumour, in which fibroblasts accumulate predominantly in the pre-neoplastic fibrotic liver and regulate the risk for hepatocellular carcinoma development through a balance of tumour-suppressive and tumour-promoting mediators. By contrast, cholangiocarcinoma is desmoplastic, with cancer-associated fibroblasts contributing to tumour growth. Accordingly, restoring the balance from tumour-promoting to tumour-suppressive fibroblasts and mediators might represent a strategy for hepatocellular carcinoma prevention, whereas in cholangiocarcinoma, fibroblasts and their mediators could be leveraged for tumour treatment. Importantly, fibroblast mediators regulating hepatocellular carcinoma development might exert opposite effects on cholangiocarcinoma growth. This Review translates the improved understanding of tumour-specific, location-specific, and stage-specific roles of fibroblasts and their mediators in liver cancer into novel and rational therapeutic concepts.
Collapse
Affiliation(s)
- Silvia Affo
- Department of Liver, Digestive System, and Metabolism, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Aveline Filliol
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Gregory J Gores
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | | |
Collapse
|
11
|
Jiang G, Wang B, Wang Y, Kong H, Wang Y, Gao P, Guo M, Li W, Zhang J, Wang Z, Niu J. Structural characteristics of a novel Bletilla striata polysaccharide and its activities for the alleviation of liver fibrosis. Carbohydr Polym 2023; 313:120781. [PMID: 37182941 DOI: 10.1016/j.carbpol.2023.120781] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 02/26/2023] [Accepted: 02/28/2023] [Indexed: 03/19/2023]
Abstract
Liver fibrosis has proven to be the main predisposing factor for liver cirrhosis and liver cancer; however, an effective treatment remains elusive. Polysaccharides, with low toxicity and a wide range of bioactivities, are strong potential candidates for anti-hepatic fibrosis applications. For this study, a new low molecular weight neutral polysaccharide (B. striata glucomannan (BSP)) was extracted and purified from Bletilla striata. The structure of BSP was characterized and its activities for alleviating liver fibrosis in vivo were further evaluated. The results revealed that the structural unit of BSP was likely →4)-β-D-Glcp-(1 → 4)-β-D-Manp-(1 → 4)-β-D-2ace-Manp-(1 → 4)-β-D-Manp-(1 → 4)-β-D-Glcp-(1 → 4)-β-D-Manp-(1 → 4)-β-D-Manp-(1 → 4)-β-D-3ace-Manp-(1→, with a molecular weight of only 58.5 kDa. Additionally, BSP was observed to attenuate the passive impacts of liver fibrosis in a manner closely related to TLR2/TLR4-MyD88-NF-κB signaling pathway conduction. In summary, the results of this study provide theoretical foundations for the potential applications of BSP as an anti-liver fibrosis platform.
Collapse
|
12
|
Hsu KS, Dunleavey JM, Szot C, Yang L, Hilton MB, Morris K, Seaman S, Feng Y, Lutz EM, Koogle R, Tomassoni-Ardori F, Saha S, Zhang XM, Zudaire E, Bajgain P, Rose J, Zhu Z, Dimitrov DS, Cuttitta F, Emenaker NJ, Tessarollo L, St. Croix B. Cancer cell survival depends on collagen uptake into tumor-associated stroma. Nat Commun 2022; 13:7078. [PMID: 36400786 PMCID: PMC9674701 DOI: 10.1038/s41467-022-34643-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 11/01/2022] [Indexed: 11/19/2022] Open
Abstract
Collagen I, the most abundant protein in humans, is ubiquitous in solid tumors where it provides a rich source of exploitable metabolic fuel for cancer cells. While tumor cells were unable to exploit collagen directly, here we show they can usurp metabolic byproducts of collagen-consuming tumor-associated stroma. Using genetically engineered mouse models, we discovered that solid tumor growth depends upon collagen binding and uptake mediated by the TEM8/ANTXR1 cell surface protein in tumor-associated stroma. Tumor-associated stromal cells processed collagen into glutamine, which was then released and internalized by cancer cells. Under chronic nutrient starvation, a condition driven by the high metabolic demand of tumors, cancer cells exploited glutamine to survive, an effect that could be reversed by blocking collagen uptake with TEM8 neutralizing antibodies. These studies reveal that cancer cells exploit collagen-consuming stromal cells for survival, exposing an important vulnerability across solid tumors with implications for developing improved anticancer therapy.
Collapse
Affiliation(s)
- Kuo-Sheng Hsu
- grid.48336.3a0000 0004 1936 8075Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI), NIH, Frederick, MD 21702 USA
| | - James M. Dunleavey
- grid.48336.3a0000 0004 1936 8075Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI), NIH, Frederick, MD 21702 USA
| | - Christopher Szot
- grid.48336.3a0000 0004 1936 8075Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI), NIH, Frederick, MD 21702 USA
| | - Liping Yang
- grid.48336.3a0000 0004 1936 8075Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI), NIH, Frederick, MD 21702 USA
| | - Mary Beth Hilton
- grid.48336.3a0000 0004 1936 8075Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI), NIH, Frederick, MD 21702 USA ,grid.418021.e0000 0004 0535 8394Basic Research Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research (FNLCR), Frederick, MD 21702 USA
| | - Karen Morris
- grid.48336.3a0000 0004 1936 8075Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI), NIH, Frederick, MD 21702 USA ,grid.418021.e0000 0004 0535 8394Basic Research Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research (FNLCR), Frederick, MD 21702 USA
| | - Steven Seaman
- grid.48336.3a0000 0004 1936 8075Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI), NIH, Frederick, MD 21702 USA
| | - Yang Feng
- grid.48336.3a0000 0004 1936 8075Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI), NIH, Frederick, MD 21702 USA
| | - Emily M. Lutz
- grid.48336.3a0000 0004 1936 8075Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI), NIH, Frederick, MD 21702 USA
| | - Robert Koogle
- grid.418021.e0000 0004 0535 8394MCGP, NCI, Frederick, MD 21702 USA
| | | | - Saurabh Saha
- BioMed Valley Discoveries, Inc, Kansas City, MO 64111 USA ,Present Address: Centessa Pharmaceuticals, Cambridge, MA 02139 USA
| | - Xiaoyan M. Zhang
- BioMed Valley Discoveries, Inc, Kansas City, MO 64111 USA ,Present Address: Ikena Oncology, Cambridge, MA 02210 USA
| | - Enrique Zudaire
- grid.48336.3a0000 0004 1936 8075Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI), NIH, Frederick, MD 21702 USA ,Present Address: Janssen Pharmaceutical Companies, J&J, R&D, Welsh Road McKean Road, Spring House, PA 19477 USA
| | - Pradip Bajgain
- grid.48336.3a0000 0004 1936 8075Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI), NIH, Frederick, MD 21702 USA
| | - Joshua Rose
- grid.48336.3a0000 0004 1936 8075Biomolecular Structure Section, Center for Structural Biology, NCI, NIH, Frederick, MD 21702 USA
| | - Zhongyu Zhu
- grid.48336.3a0000 0004 1936 8075Protein Interactions Section, Cancer and Inflammation Program, NCI, NIH, Frederick, MD 21702 USA ,grid.420872.bPresent Address: Lentigen Technology, Inc. 1201 Clopper Road, Gaithersburg, MD 20878 USA
| | - Dimiter S. Dimitrov
- grid.48336.3a0000 0004 1936 8075Protein Interactions Section, Cancer and Inflammation Program, NCI, NIH, Frederick, MD 21702 USA ,grid.21925.3d0000 0004 1936 9000Present Address: Center for Antibody Therapeutics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261 USA
| | - Frank Cuttitta
- grid.48336.3a0000 0004 1936 8075Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI), NIH, Frederick, MD 21702 USA
| | - Nancy J. Emenaker
- grid.48336.3a0000 0004 1936 8075Division of Cancer Prevention, NCI, NIH, Bethesda, MD 20892 USA
| | - Lino Tessarollo
- grid.48336.3a0000 0004 1936 8075Neural Development Section, MCGP, NCI, NIH, Frederick, MD 21702 USA
| | - Brad St. Croix
- grid.48336.3a0000 0004 1936 8075Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI), NIH, Frederick, MD 21702 USA
| |
Collapse
|
13
|
Filliol A, Saito Y, Nair A, Dapito DH, Yu LX, Ravichandra A, Bhattacharjee S, Affo S, Fujiwara N, Su H, Sun Q, Savage TM, Wilson-Kanamori JR, Caviglia JM, Chin L, Chen D, Wang X, Caruso S, Kang JK, Amin AD, Wallace S, Dobie R, Yin D, Rodriguez-Fiallos OM, Yin C, Mehal A, Izar B, Friedman RA, Wells RG, Pajvani UB, Hoshida Y, Remotti HE, Arpaia N, Zucman-Rossi J, Karin M, Henderson NC, Tabas I, Schwabe RF. Opposing roles of hepatic stellate cell subpopulations in hepatocarcinogenesis. Nature 2022; 610:356-365. [PMID: 36198802 PMCID: PMC9949942 DOI: 10.1038/s41586-022-05289-6] [Citation(s) in RCA: 182] [Impact Index Per Article: 60.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 08/30/2022] [Indexed: 01/21/2023]
Abstract
Hepatocellular carcinoma (HCC), the fourth leading cause of cancer mortality worldwide, develops almost exclusively in patients with chronic liver disease and advanced fibrosis1,2. Here we interrogated functions of hepatic stellate cells (HSCs), the main source of liver fibroblasts3, during hepatocarcinogenesis. Genetic depletion, activation or inhibition of HSCs in mouse models of HCC revealed their overall tumour-promoting role. HSCs were enriched in the preneoplastic environment, where they closely interacted with hepatocytes and modulated hepatocarcinogenesis by regulating hepatocyte proliferation and death. Analyses of mouse and human HSC subpopulations by single-cell RNA sequencing together with genetic ablation of subpopulation-enriched mediators revealed dual functions of HSCs in hepatocarcinogenesis. Hepatocyte growth factor, enriched in quiescent and cytokine-producing HSCs, protected against hepatocyte death and HCC development. By contrast, type I collagen, enriched in activated myofibroblastic HSCs, promoted proliferation and tumour development through increased stiffness and TAZ activation in pretumoural hepatocytes and through activation of discoidin domain receptor 1 in established tumours. An increased HSC imbalance between cytokine-producing HSCs and myofibroblastic HSCs during liver disease progression was associated with increased HCC risk in patients. In summary, the dynamic shift in HSC subpopulations and their mediators during chronic liver disease is associated with a switch from HCC protection to HCC promotion.
Collapse
Affiliation(s)
- Aveline Filliol
- Department of Medicine, Columbia University, New York, NY, USA
| | - Yoshinobu Saito
- Department of Medicine, Columbia University, New York, NY, USA
| | - Ajay Nair
- Department of Medicine, Columbia University, New York, NY, USA
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Dianne H Dapito
- Department of Medicine, Columbia University, New York, NY, USA
| | - Le-Xing Yu
- Department of Medicine, Columbia University, New York, NY, USA
| | - Aashreya Ravichandra
- Department of Medicine, Columbia University, New York, NY, USA
- Klinikum Rechts der Isar, Technical University of Munich (TUM), Munich, Germany
| | | | - Silvia Affo
- Department of Medicine, Columbia University, New York, NY, USA
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Naoto Fujiwara
- Liver Tumor Translational Research Program, Harold C. Simmons Comprehensive Cancer Center, Division of Digestive and Liver Diseases, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Hua Su
- Department of Pharmacology, School of Medicine, University of California, San Diego, San Diego, CA, USA
| | - Qiuyan Sun
- Department of Medicine, Columbia University, New York, NY, USA
| | - Thomas M Savage
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - John R Wilson-Kanamori
- Centre for Inflammation Research, The Queen's Medical Research Institute, Edinburgh BioQuarter, University of Edinburgh, Edinburgh, UK
| | - Jorge M Caviglia
- Department of Medicine, Columbia University, New York, NY, USA
- Department of Health and Nutrition Sciences, Brooklyn College, City University of New York, New York, NY, USA
| | - LiKang Chin
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Biomedical Engineering, Widener University, Chester, PA, USA
| | - Dongning Chen
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Xiaobo Wang
- Department of Medicine, Columbia University, New York, NY, USA
| | - Stefano Caruso
- Functional Genomics of Solid Tumors Laboratory, Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Paris, France
| | - Jin Ku Kang
- Department of Medicine, Columbia University, New York, NY, USA
- Institute of Human Nutrition, Columbia University, New York, NY, USA
| | - Amit Dipak Amin
- Department of Medicine, Columbia University, New York, NY, USA
| | - Sebastian Wallace
- Centre for Inflammation Research, The Queen's Medical Research Institute, Edinburgh BioQuarter, University of Edinburgh, Edinburgh, UK
| | - Ross Dobie
- Centre for Inflammation Research, The Queen's Medical Research Institute, Edinburgh BioQuarter, University of Edinburgh, Edinburgh, UK
| | - Deqi Yin
- Department of Medicine, Columbia University, New York, NY, USA
| | | | - Chuan Yin
- Department of Medicine, Columbia University, New York, NY, USA
- Department of Gastroenterology, Changzheng Hospital, Shanghai, China
| | - Adam Mehal
- Department of Medicine, Columbia University, New York, NY, USA
| | - Benjamin Izar
- Department of Medicine, Columbia University, New York, NY, USA
| | - Richard A Friedman
- Biomedical Informatics Shared Resource, Herbert Irving Comprehensive Cancer Center, and Department of Biomedical Informatics, Columbia University Irving Medical Center, New York, NY, USA
| | - Rebecca G Wells
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Utpal B Pajvani
- Department of Medicine, Columbia University, New York, NY, USA
- Institute of Human Nutrition, Columbia University, New York, NY, USA
| | - Yujin Hoshida
- Liver Tumor Translational Research Program, Harold C. Simmons Comprehensive Cancer Center, Division of Digestive and Liver Diseases, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Helen E Remotti
- Department of Pathology, Columbia University Irving Medical Center, New York, NY, USA
| | - Nicholas Arpaia
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Jessica Zucman-Rossi
- Functional Genomics of Solid Tumors Laboratory, Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Paris, France
| | - Michael Karin
- Department of Pharmacology, School of Medicine, University of California, San Diego, San Diego, CA, USA
| | - Neil C Henderson
- Centre for Inflammation Research, The Queen's Medical Research Institute, Edinburgh BioQuarter, University of Edinburgh, Edinburgh, UK
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Ira Tabas
- Department of Medicine, Columbia University, New York, NY, USA
- Institute of Human Nutrition, Columbia University, New York, NY, USA
- Department of Pathology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Physiology, Columbia University, New York, NY, USA
| | - Robert F Schwabe
- Department of Medicine, Columbia University, New York, NY, USA.
- Institute of Human Nutrition, Columbia University, New York, NY, USA.
| |
Collapse
|
14
|
Garcia-Arranz M, Villarejo-Campos P, Barambio J, Garcia Gomez-Heras S, Vega-Clemente L, Guadalajara H, García-Olmo D. Toxicity study in a pig model of intraperitoneal collagenase as an "enzymatic scalpel" directed to break stroma in order to generate a new perspective for peritoneal carcinomatosis approach: an experimental research. World J Surg Oncol 2022; 20:53. [PMID: 35216593 PMCID: PMC8881860 DOI: 10.1186/s12957-022-02524-2] [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: 10/20/2021] [Accepted: 02/05/2022] [Indexed: 11/22/2022] Open
Abstract
Background This study aimed to measure the toxicity resulting from collagenase administration to the peritoneal cavity in a pig model as a preliminary step to break down the stroma surrounding tumors. Methods Eight pigs were treated with 2 different collagenase concentrations previously tested in rats by our group. Time and temperature were controlled using a peritoneal lavage system (PRS System, Combat Medical Ltd.) identical to that used in human surgeries through hyperthermic intraperitoneal chemotherapy (HIPEC); 2 additional pigs were treated with peritoneal lavage only. Samples of blood and peritoneal fluid were collected pre-treatment, immediately after treatment, and 24 h postoperatively. In addition, histological studies and blood collagenase levels were measured. Results No complications were observed during the surgeries. Intraoperative images evidenced the release of peritoneal tissue during collagenase treatment. After surgery, the animals showed no signs of pain. Diet and mobility were normal at 4 h postoperatively, and there were no significant differences in hematologic or biochemical parameters. Quantification of MMP1 and MMP2 in all samples as measured by absorbance showed no differences in blood collagenase levels between pre-treatment, post-treatment, and 24 h postoperatively. None of the animals treated with collagenase showed peritoneal adhesions during the second surgery. Histologically, peritoneal organs and serous structures did not show any microscopic alterations associated with collagenase treatment in any group. Conclusion Lavage of the peritoneal cavity with doses of up to 100,000 collagen digestion units/animal for 30 min is safe and removes connective tissue from the peritoneal cavity.
Collapse
Affiliation(s)
- M Garcia-Arranz
- New Therapies Laboratory, Health Research Institute-Fundación Jiménez Díaz University Hospital (IIS-FJD), Avda. Reyes Católicos, 2, 28040, Madrid, Spain. .,Department of Surgery, Universidad Autónoma de Madrid, C/ Arzobispo Morcillo s/n, 28034, Madrid, Spain.
| | - P Villarejo-Campos
- Department of Surgery, Fundación Jiménez Díaz University Hospital, Avda. Reyes Católicos, 2, 28040, Madrid, Spain
| | - J Barambio
- Department of Surgery, Fundación Jiménez Díaz University Hospital, Avda. Reyes Católicos, 2, 28040, Madrid, Spain
| | - S Garcia Gomez-Heras
- Department of Human Histology, Universidad Rey Juan Carlos, Avda de Atenas s/n, 28922, Alcorcón, Spain
| | - L Vega-Clemente
- New Therapies Laboratory, Health Research Institute-Fundación Jiménez Díaz University Hospital (IIS-FJD), Avda. Reyes Católicos, 2, 28040, Madrid, Spain
| | - H Guadalajara
- Department of Surgery, Universidad Autónoma de Madrid, C/ Arzobispo Morcillo s/n, 28034, Madrid, Spain.,Department of Surgery, Fundación Jiménez Díaz University Hospital, Avda. Reyes Católicos, 2, 28040, Madrid, Spain
| | - D García-Olmo
- New Therapies Laboratory, Health Research Institute-Fundación Jiménez Díaz University Hospital (IIS-FJD), Avda. Reyes Católicos, 2, 28040, Madrid, Spain.,Department of Surgery, Universidad Autónoma de Madrid, C/ Arzobispo Morcillo s/n, 28034, Madrid, Spain.,Department of Surgery, Fundación Jiménez Díaz University Hospital, Avda. Reyes Católicos, 2, 28040, Madrid, Spain
| |
Collapse
|
15
|
Rømer AMA, Thorseth ML, Madsen DH. Immune Modulatory Properties of Collagen in Cancer. Front Immunol 2021; 12:791453. [PMID: 34956223 PMCID: PMC8692250 DOI: 10.3389/fimmu.2021.791453] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 11/22/2021] [Indexed: 12/22/2022] Open
Abstract
During tumor growth the extracellular matrix (ECM) undergoes dramatic remodeling. The normal ECM is degraded and substituted with a tumor-specific ECM, which is often of higher collagen density and increased stiffness. The structure and collagen density of the tumor-specific ECM has been associated with poor prognosis in several types of cancer. However, the reason for this association is still largely unknown. Collagen can promote cancer cell growth and migration, but recent studies have shown that collagens can also affect the function and phenotype of various types of tumor-infiltrating immune cells such as tumor-associated macrophages (TAMs) and T cells. This suggests that tumor-associated collagen could have important immune modulatory functions within the tumor microenvironment, affecting cancer progression as well as the efficacy of cancer immunotherapy. The effects of tumor-associated collagen on immune cells could help explain why a high collagen density in tumors is often correlated with a poor prognosis. Knowledge about immune modulatory functions of collagen could potentially identify targets for improving current cancer therapies or for development of new treatments. In this review, the current knowledge about the ability of collagen to influence T cell activity will be summarized. This includes direct interactions with T cells as well as induction of immune suppressive activity in other immune cells such as macrophages. Additionally, the potential effects of collagen on the efficacy of cancer immunotherapy will be discussed.
Collapse
Affiliation(s)
- Anne Mette Askehøj Rømer
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark.,Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | - Marie-Louise Thorseth
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark.,Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Daniel Hargbøl Madsen
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark.,Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|