1
|
Germitsch N, Kockmann T, Schnyder M, Tritten L. N-terminomics profiling of host proteins targeted by excretory-secretory proteases of the nematode Angiostrongylus vasorum identifies points of interaction with canine coagulation and complement cascade. PLoS One 2025; 20:e0316217. [PMID: 39813225 PMCID: PMC11734930 DOI: 10.1371/journal.pone.0316217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 12/07/2024] [Indexed: 01/18/2025] Open
Abstract
The cardiopulmonary nematode Angiostrongylus vasorum can cause severe disease in dogs, including coagulopathies manifesting with bleeding. We analysed A. vasorum excretory/secretory protein (ESP)-treated dog plasma and serum by N-terminome analysis using Terminal Amine Isotopic Labelling of Substrates (TAILS) to identify cleaved host substrates. In plasma and serum samples 430 and 475 dog proteins were identified, respectively. A total of eight dog proteins were significantly cleaved at higher levels upon exposure to A. vasorum ESP: of these, three were coagulation factors (factor II, V and IX) and three were complement proteins (complement C3, C4-A and C5). Comparison with human motif sequence orthologues revealed known cleavage sites in coagulation factor IX and II (prothrombin). These and further identified cleavage sites suggest direct or indirect activation or proteolysis of complement and coagulation components through A. vasorum ESP, which contains several proteases. Further studies are needed to validate their substrate specificity.
Collapse
Affiliation(s)
- Nina Germitsch
- Institute of Parasitology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Tobias Kockmann
- Functional Genomics Center Zurich, Swiss Federal Institute of Technology Zurich (ETH Zurich)/ University of Zurich, Zurich, Switzerland
| | - Manuela Schnyder
- Institute of Parasitology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Lucienne Tritten
- Institute of Parasitology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| |
Collapse
|
2
|
Desroys du Roure P, David T, Mallavialle A, Laurent-Matha V, Roger P, Guiu S, Chardès T, Liaudet-Coopman E. Antibodies against the multifaceted cathepsin D protein open new avenues for TNBC immunotherapy. J Immunother Cancer 2025; 13:e009548. [PMID: 39800383 PMCID: PMC11748927 DOI: 10.1136/jitc-2024-009548] [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: 04/26/2024] [Accepted: 12/02/2024] [Indexed: 01/23/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is a heterogeneous breast cancer subtype characterized by aggressive clinical behavior and poor prognosis. The immune landscape associated with TNBC often reveals high immunogenicity. Therefore, immunotherapy, which has demonstrated its efficacy in different cancer types, could be a promising strategy for TNBC, given the limited therapeutic options currently available besides conventional chemotherapy. The aspartic protease cathepsin D (cath-D) is a tumor cell-associated extracellular protein with protumor activity, a marker of poor prognosis, and a target for antibody-based therapy in TNBC. This commentary provides a synopsis/narrative summary of the development of anti-cath-D antibodies in different formats, their key roles in restoring the antitumor immunity, particularly via activation of tumor-infiltrating natural killer cells, and their dual antitumor effects on cancer cells and stromal cancer-associated fibroblasts, suggesting their interest for clinical use in the light of the current clinical knowledge on TNBC.
Collapse
Affiliation(s)
| | - Timothée David
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
| | - Aude Mallavialle
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
| | | | - Pascal Roger
- Department of Pathology, CHU Nîmes, Nîmes, France
| | - Séverine Guiu
- Department of Medical Oncology, ICM, Montpellier, France
| | - Thierry Chardès
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
- CNRS, Centre National de la Recherche Scientifique, Paris, F-75016, France
| | | |
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
|
Xiao G, Wei Y, Xie R, Tsang Y, Gu J, Shen D, Ding M, Yuan J, Xu D, Fei J. Citric acid promotes SPARC release in pancreatic cancer cells and inhibits the progression of pancreatic tumors in mice on a high-fat diet. FEBS J 2024; 291:1699-1718. [PMID: 38245817 DOI: 10.1111/febs.17058] [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: 06/20/2023] [Revised: 10/17/2023] [Accepted: 01/08/2024] [Indexed: 01/22/2024]
Abstract
Over the years, pancreatic cancer has experienced a global surge in incidence and mortality rates, largely attributed to the influence of obesity and diabetes mellitus on disease initiation and progression. In this study, we investigated the pathogenesis of pancreatic cancer in mice subjected to a high-fat diet (HFD) and observed an increase in citric acid expenditure. Notably, citrate treatment demonstrates significant efficacy in promoting tumor cell apoptosis, suppressing cell proliferation, and inhibiting tumor growth in vivo. Our investigations revealed that citrate achieved these effects by releasing secreted protein acidic and rich in cysteine (SPARC) proteins, repolarizing M2 macrophages into M1 macrophages, and facilitating tumor cell apoptosis. Overall, our research highlights the critical role of citric acid as a pivotal metabolite in the intricate relationship between obesity and pancreatic cancer. Furthermore, we uncovered the significant metabolic and immune checkpoint function of SPARC in pancreatic cancer, suggesting its potential as both a biomarker and therapeutic target in treating this patient population.
Collapse
Affiliation(s)
- Guohui Xiao
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Yan Wei
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, China
| | - Rongli Xie
- Department of General Surgery, Ruijin Hospital Luwan Branch, Shanghai Jiao Tong University School of Medicine, China
| | - Yiusing Tsang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Jianhua Gu
- Department of Thyroid and Breast Surgery, Punan Branch of Renji Hospital, Shanghai Jiaotong University School of Medicine, China
| | - Dongjie Shen
- Department of General Surgery, Ruijin Hospital Luwan Branch, Shanghai Jiao Tong University School of Medicine, China
| | - Min Ding
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Jianming Yuan
- Department of General Surgery, Ruijin Hospital Luwan Branch, Shanghai Jiao Tong University School of Medicine, China
| | - Dan Xu
- Department of Emergency Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Jian Fei
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, China
- State Key Laboratory of Oncogenes and Related Genes (Shanghai), China
- Institute of Translational Medicine, Shanghai Jiao Tong University, China
| |
Collapse
|
5
|
Desroys du Roure P, Lajoie L, Mallavialle A, Alcaraz LB, Mansouri H, Fenou L, Garambois V, Rubio L, David T, Coenon L, Boissière-Michot F, Chateau MC, Ngo G, Jarlier M, Villalba M, Martineau P, Laurent-Matha V, Roger P, Guiu S, Chardès T, Gros L, Liaudet-Coopman E. A novel Fc-engineered cathepsin D-targeting antibody enhances ADCC, triggers tumor-infiltrating NK cell recruitment, and improves treatment with paclitaxel and enzalutamide in triple-negative breast cancer. J Immunother Cancer 2024; 12:e007135. [PMID: 38290768 PMCID: PMC10828871 DOI: 10.1136/jitc-2023-007135] [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] [Accepted: 12/19/2023] [Indexed: 02/01/2024] Open
Abstract
INTRODUCTION Triple-negative breast cancer (TNBC) prognosis is poor. Immunotherapies to enhance the antibody-induced natural killer (NK) cell antitumor activity are emerging for TNBC that is frequently immunogenic. The aspartic protease cathepsin D (cath-D), a tumor cell-associated extracellular protein with protumor activity and a poor prognosis marker in TNBC, is a prime target for antibody-based therapy to induce NK cell-mediated antibody-dependent cellular cytotoxicity (ADCC). This study investigated whether Fc-engineered anti-cath-D antibodies trigger ADCC, their impact on antitumor efficacy and tumor-infiltrating NK cells, and their relevance for combinatory therapy in TNBC. METHODS Cath-D expression and localization in TNBC samples were evaluated by western blotting, immunofluorescence, and immunohistochemistry. The binding of human anti-cath-D F1M1 and Fc-engineered antibody variants, which enhance (F1M1-Fc+) or prevent (F1M1-Fc-) affinity for CD16a, to secreted human and murine cath-D was analyzed by ELISA, and to CD16a by surface plasmon resonance and flow cytometry. NK cell activation was investigated by flow cytometry, and ADCC by lactate dehydrogenase release. The antitumor efficacy of F1M1 Fc-variants was investigated using TNBC cell xenografts in nude mice. NK cell recruitment, activation, and cytotoxic activity were analyzed in MDA-MB-231 cell xenografts by immunophenotyping and RT-qPCR. NK cells were depleted using an anti-asialo GM1 antibody. F1M1-Fc+ antitumor effect was assessed in TNBC patient-derived xenografts (PDXs) and TNBC SUM159 cell xenografts, and in combination with paclitaxel or enzalutamide. RESULTS Cath-D expression on the TNBC cell surface could be exploited to induce ADCC. F1M1 Fc-variants recognized human and mouse cath-D. F1M1-Fc+ activated NK cells in vitro and induced ADCC against TNBC cells and cancer-associated fibroblasts more efficiently than F1M1. F1M1-Fc- was ineffective. In the MDA-MB-231 cell xenograft model, F1M1-Fc+ displayed higher antitumor activity than F1M1, whereas F1M1-Fc- was less effective, reflecting the importance of Fc-dependent mechanisms in vivo. F1M1-Fc+ triggered tumor-infiltrating NK cell recruitment, activation and cytotoxic activity in MDA-MB-231 cell xenografts. NK cell depletion impaired F1M1-Fc+ antitumor activity, demonstrating their key role. F1M1-Fc+ inhibited growth of SUM159 cell xenografts and two TNBC PDXs. In combination therapy, F1M1-Fc+ improved paclitaxel and enzalutamide therapeutic efficacy without toxicity. CONCLUSIONS F1M1-Fc+ is a promising immunotherapy for TNBC that could be combined with conventional regimens, including chemotherapy or antiandrogens.
Collapse
Affiliation(s)
| | - Laurie Lajoie
- Université de Tours - INRAE, UMR1282, Infectiologie et Santé Publique (ISP), équipe BioMédicaments Anti-Parasitaires (BioMAP), Tours, France
| | - Aude Mallavialle
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
| | - Lindsay B Alcaraz
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
| | - Hanane Mansouri
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
- RHEM, IRCM, Montpellier, France
| | - Lise Fenou
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
| | | | - Lucie Rubio
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
| | - Timothée David
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
| | - Loïs Coenon
- IRMB, University of Montpellier, INSERM, CNRS, CHU Montpellier, Montpellier, France
| | | | | | - Giang Ngo
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
| | | | - Martin Villalba
- IRMB, University of Montpellier, INSERM, CNRS, CHU Montpellier, Montpellier, France
- Institut du Cancer Avignon-Provence Sainte Catherine, Avignon, France
| | - Pierre Martineau
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
| | | | - Pascal Roger
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
- Department of Pathology, CHU Nîmes, Nimes, France
| | - Séverine Guiu
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
- Department of Medical Oncology, ICM, Montpellier, France
| | - Thierry Chardès
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
- CNRS, Centre national de la recherche Scientifique, Paris, F-75016, France
| | - Laurent Gros
- IRCM, INSERM U1194, University of Montpellier, ICM, Montpellier, France
- CNRS, Centre national de la recherche Scientifique, Paris, F-75016, France
| | | |
Collapse
|
6
|
David T, Mallavialle A, Faget J, Alcaraz LB, Lapierre M, du Roure PD, Laurent-Matha V, Mansouri H, Jarlier M, Martineau P, Roger P, Guiu S, Chardès T, Liaudet-Coopman E. Anti-cathepsin D immunotherapy triggers both innate and adaptive anti-tumour immunity in breast cancer. Br J Pharmacol 2023. [PMID: 38030588 DOI: 10.1111/bph.16291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 11/13/2023] [Accepted: 11/17/2023] [Indexed: 12/01/2023] Open
Abstract
BACKGROUND AND PURPOSE Triple-negative breast cancer (TNBC) has poorer outcomes than other breast cancers (BC), including HER2+ BC. Cathepsin D (CathD) is a poor prognosis marker overproduced by BC cells, hypersecreted in the tumour microenvironment with tumour-promoting activity. Here, we characterized the immunomodulatory activity of the anti-CathD antibody F1 and its improved Fab-aglycosylated version (F1M1) in immunocompetent mouse models of TNBC (C57BL/6 mice harbouring E0771 cell grafts) and HER2-amplified BC (BALB/c mice harbouring TUBO cell grafts). EXPERIMENTAL APPROACH CathD expression was evaluated by western blotting and immunofluorescence, and antibody binding to CathD by ELISA. Antibody anti-tumour efficacy was investigated in mouse models. Immune cell recruitment and activation were assessed by immunohistochemistry, immunophenotyping, and RT-qPCR. KEY RESULTS F1 and F1M1 antibodies remodelled the tumour immune landscape. Both antibodies promoted innate antitumour immunity by preventing the recruitment of immunosuppressive M2-polarized tumour-associated macrophages (TAMs) and by activating natural killer cells in the tumour microenvironment of both models. This translated into a reduction of T-cell exhaustion markers in the tumour microenvironment that could be locally supported by enhanced activation of anti-tumour antigen-presenting cell (M1-polarized TAMs and cDC1 cells) functions. Both antibodies inhibited tumour growth in the highly-immunogenic E0771 model, but only marginally in the immune-excluded TUBO model, indicating that anti-CathD immunotherapy is more relevant for BC with a high immune cell infiltrate, as often observed in TNBC. CONCLUSION AND IMPLICATION Anti-CathD antibody-based therapy triggers the anti-tumour innate and adaptive immunity in preclinical models of BC and is a promising immunotherapy for immunogenic TNBC.
Collapse
Affiliation(s)
- Timothée David
- IRCM, INSERM U1194, Univ Montpellier, ICM, Montpellier, France
| | | | - Julien Faget
- IRCM, INSERM U1194, Univ Montpellier, ICM, Montpellier, France
| | | | - Marion Lapierre
- IRCM, INSERM U1194, Univ Montpellier, ICM, Montpellier, France
| | | | | | - Hanane Mansouri
- IRCM, INSERM U1194, Univ Montpellier, ICM, Montpellier, France
- RHEM, IRCM, Montpellier, France
| | | | | | - Pascal Roger
- IRCM, INSERM U1194, Univ Montpellier, ICM, Montpellier, France
- Department of Pathology, CHU Nîmes, Nîmes, France
| | - Séverine Guiu
- IRCM, INSERM U1194, Univ Montpellier, ICM, Montpellier, France
- Department of Medical Oncology, ICM, Montpellier, France
| | - Thierry Chardès
- IRCM, INSERM U1194, Univ Montpellier, ICM, Montpellier, France
- Centre national de la recherche Scientifique, CNRS, Paris, France
| | | |
Collapse
|
7
|
Wang G, Zhang H, Shen X, Jin W, Wang X, Zhou Z. Characterization of cancer-associated fibroblasts (CAFs) and development of a CAF-based risk model for triple-negative breast cancer. Cancer Cell Int 2023; 23:294. [PMID: 38007443 PMCID: PMC10676599 DOI: 10.1186/s12935-023-03152-w] [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: 07/08/2023] [Accepted: 11/19/2023] [Indexed: 11/27/2023] Open
Abstract
Triple-negative breast Cancer (TNBC) is a highly malignant cancer with unclear pathogenesis. Within the tumor microenvironment (TME), cancer-associated fibroblasts (CAFs) vitally influence tumor onset and progression. Thus, this research aimed to identify distinct subgroups of CAF using single-cell and TNBC-related information from the GEO and TCGA databases, respectively. The primary aim was to establish a novel predictive model based on the CAF features and their clinical relevance. Moreover, the CAFs were analyzed for their immune characteristics, response to immunotherapy, and sensitivity to different drugs. The developed predictive model demonstrated significant effectiveness in determining the prognosis of patients with TNBC, TME, and the immune landscape of the tumor. Of note, the expression of GPR34 was significantly higher in TNBC tissues compared to that in other breast cancer (non-TNBC) tissues, indicating that GPR34 plays a crucial role in the onset and progression of TNBC. In summary, this research has yielded a novel predictive model for TNBC that holds promise for the accurate prediction of prognosis and response to immunotherapy in patients with TNBC.
Collapse
Affiliation(s)
- Ganggang Wang
- Department of Hepatobiliary Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Hao Zhang
- Department of Hepatobiliary Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Xiaowei Shen
- Department of General Surgery, Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wenzhi Jin
- Department of Hepatobiliary Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Xiaoliang Wang
- Department of Hepatobiliary Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China.
| | - Zhijie Zhou
- Department of Hepatobiliary Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China.
| |
Collapse
|
8
|
Jiang S, Sun HF, Li S, Zhang N, Chen JS, Liu JX. SPARC: a potential target for functional nanomaterials and drugs. Front Mol Biosci 2023; 10:1235428. [PMID: 37577749 PMCID: PMC10419254 DOI: 10.3389/fmolb.2023.1235428] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 07/13/2023] [Indexed: 08/15/2023] Open
Abstract
Secreted protein acidic and rich in cysteine (SPARC), also termed osteonectin or BM-40, is a matricellular protein which regulates cell adhesion, extracellular matrix production, growth factor activity, and cell cycle. Although SPARC does not perform a structural function, it, however, modulates interactions between cells and the surrounding extracellular matrix due to its anti-proliferative and anti-adhesion properties. The overexpression of SPARC at sites, including injury, regeneration, obesity, cancer, and inflammation, reveals its application as a prospective target and therapeutic indicator in the treatment and assessment of disease. This article comprehensively summarizes the mechanism of SPARC overexpression in inflammation and tumors as well as the latest research progress of functional nanomaterials in the therapy of rheumatoid arthritis and tumors by manipulating SPARC as a new target. This article provides ideas for using functional nanomaterials to treat inflammatory diseases through the SPARC target. The purpose of this article is to provide a reference for ongoing disease research based on SPARC-targeted therapy.
Collapse
Affiliation(s)
- Shan Jiang
- School of Pharmacy, Heilongjiang University of Traditional Chinese Medicine, Harbin, China
- School of Pharmaceutical Sciences, Department of Rehabilitation and Healthcare, Hunan University of Medicine, Huaihua, China
| | - Hui-Feng Sun
- School of Pharmacy, Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Shuang Li
- School of Pharmaceutical Sciences, Department of Rehabilitation and Healthcare, Hunan University of Medicine, Huaihua, China
- College Pharmacy, Jiamusi University, Jiamusi, China
| | - Ning Zhang
- School of Pharmacy, Heilongjiang University of Traditional Chinese Medicine, Harbin, China
- School of Pharmaceutical Sciences, Department of Rehabilitation and Healthcare, Hunan University of Medicine, Huaihua, China
| | - Ji-Song Chen
- School of Pharmaceutical Sciences, Department of Rehabilitation and Healthcare, Hunan University of Medicine, Huaihua, China
| | - Jian-Xin Liu
- School of Pharmaceutical Sciences, Department of Rehabilitation and Healthcare, Hunan University of Medicine, Huaihua, China
- School of Pharmaceutical Sciences, University of South China, Hengyang, China
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
9
|
Alcaraz LB, Mallavialle A, Mollevi C, Boissière-Michot F, Mansouri H, Simony-Lafontaine J, Laurent-Matha V, Chardès T, Jacot W, Turtoi A, Roger P, Guiu S, Liaudet-Coopman E. SPARC in cancer-associated fibroblasts is an independent poor prognostic factor in non-metastatic triple-negative breast cancer and exhibits pro-tumor activity. Int J Cancer 2023; 152:1243-1258. [PMID: 36346290 PMCID: PMC10099777 DOI: 10.1002/ijc.34345] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 10/17/2022] [Accepted: 10/20/2022] [Indexed: 11/09/2022]
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive breast cancer subtype and lacks specific targeted therapeutic agents. The current mechanistic evidence from cell-based studies suggests that the matricellular protein SPARC has a tumor-promoting role in TNBC; however, data on the clinical relevance of SPARC expression/secretion by tumor and stromal cells in TNBC are limited. Here, we analyzed by immunohistochemistry the prognostic value of tumor and stromal cell SPARC expression in 148 patients with non-metastatic TNBC and long follow-up (median: 5.4 years). We also quantified PD-L1 and PD-1 expression. We detected SPARC expression in tumor cells (42.4%), cancer-associated fibroblasts (CAFs; 88.1%), tumor-associated macrophages (77.1%), endothelial cells (75.2%) and tumor-infiltrating lymphocytes (9.8%). Recurrence-free survival was significantly lower in patients with SPARC-expressing CAFs. Multivariate analysis showed that SPARC expression in CAFs was an independent prognostic factor. We also detected tumor and stromal cell SPARC expression in TNBC cytosols, and in patient-derived xenografts and cell lines. Furthermore, we analyzed publicly available single-cell mRNA sequencing data and found that in TNBC, SPARC is expressed by different CAF subpopulations, including myofibroblasts and inflammatory fibroblasts that are involved in tumor-related processes. We then showed that fibroblast-secreted SPARC had a tumor-promoting role by inhibiting TNBC cell adhesion and stimulating their motility and invasiveness. Overall, our study demonstrates that SPARC expression in CAFs is an independent prognostic marker of poor outcome in TNBC. Patients with SPARC-expressing CAFs could be eligible for anti-SPARC targeted therapy.
Collapse
Affiliation(s)
| | | | - Caroline Mollevi
- Biometry Unit, ICM, University of Montpellier, Montpellier, France.,Desbrest Institute of Epidemiology and Public Health, University of Montpellier, INSERM, Montpellier, France
| | | | - Hanane Mansouri
- IRCM, INSERM U1194, Univ Montpellier, ICM, Montpellier, France.,RHEM, IRCM, Montpellier, France
| | | | | | - Thierry Chardès
- IRCM, INSERM U1194, Univ Montpellier, ICM, Montpellier, France
| | - William Jacot
- IRCM, INSERM U1194, Univ Montpellier, ICM, Montpellier, France.,Translational Research Unit, ICM, Montpellier, France.,Department of Medical Oncology, ICM, Montpellier, France
| | - Andrei Turtoi
- IRCM, INSERM U1194, Univ Montpellier, ICM, Montpellier, France
| | - Pascal Roger
- IRCM, INSERM U1194, Univ Montpellier, ICM, Montpellier, France.,Department of Pathology, CHU, Nîmes, France
| | - Séverine Guiu
- IRCM, INSERM U1194, Univ Montpellier, ICM, Montpellier, France.,Department of Medical Oncology, ICM, Montpellier, France
| | | |
Collapse
|
10
|
Punetha A, Kotiya D. Advancements in Oncoproteomics Technologies: Treading toward Translation into Clinical Practice. Proteomes 2023; 11:2. [PMID: 36648960 PMCID: PMC9844371 DOI: 10.3390/proteomes11010002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 01/03/2023] [Accepted: 01/04/2023] [Indexed: 01/12/2023] Open
Abstract
Proteomics continues to forge significant strides in the discovery of essential biological processes, uncovering valuable information on the identity, global protein abundance, protein modifications, proteoform levels, and signal transduction pathways. Cancer is a complicated and heterogeneous disease, and the onset and progression involve multiple dysregulated proteoforms and their downstream signaling pathways. These are modulated by various factors such as molecular, genetic, tissue, cellular, ethnic/racial, socioeconomic status, environmental, and demographic differences that vary with time. The knowledge of cancer has improved the treatment and clinical management; however, the survival rates have not increased significantly, and cancer remains a major cause of mortality. Oncoproteomics studies help to develop and validate proteomics technologies for routine application in clinical laboratories for (1) diagnostic and prognostic categorization of cancer, (2) real-time monitoring of treatment, (3) assessing drug efficacy and toxicity, (4) therapeutic modulations based on the changes with prognosis and drug resistance, and (5) personalized medication. Investigation of tumor-specific proteomic profiles in conjunction with healthy controls provides crucial information in mechanistic studies on tumorigenesis, metastasis, and drug resistance. This review provides an overview of proteomics technologies that assist the discovery of novel drug targets, biomarkers for early detection, surveillance, prognosis, drug monitoring, and tailoring therapy to the cancer patient. The information gained from such technologies has drastically improved cancer research. We further provide exemplars from recent oncoproteomics applications in the discovery of biomarkers in various cancers, drug discovery, and clinical treatment. Overall, the future of oncoproteomics holds enormous potential for translating technologies from the bench to the bedside.
Collapse
Affiliation(s)
- Ankita Punetha
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Rutgers University, 225 Warren St., Newark, NJ 07103, USA
| | - Deepak Kotiya
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, 900 South Limestone St., Lexington, KY 40536, USA
| |
Collapse
|
11
|
Boissière-Michot F, Chateau MC, Thézenas S, Guiu S, Bobrie A, Jacot W. Correlation of the TIGIT-PVR immune checkpoint axis with clinicopathological features in triple-negative breast cancer. Front Immunol 2022; 13:1058424. [PMID: 36544779 PMCID: PMC9760730 DOI: 10.3389/fimmu.2022.1058424] [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: 09/30/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022] Open
Abstract
Background T cell immunoreceptor with Ig and ITIM domains (TIGIT) interacts with poliovirus receptor (PVR) to contribute to cancer immune escape. Recently, TIGIT and PVR have been identified as promising immunotherapy targets. Their gene expression is upregulated in many solid tumors, but their protein expression level is not well documented, particularly in triple negative breast cancer (TNBC), the breast cancer subtype that most benefit from immunotherapy. Methods TIGIT and PVR expression levels were assessed by immunohistochemistry in 243 surgically resected localized TNBC and then their relationship with clinical-pathological features and clinical outcome was analyzed. Results TIGIT expression was observed in immune cells from the tumor microenvironment, whereas PVR was mainly expressed by tumor cells. High TIGIT expression was significantly associated with age (p=0.010), histological grade (p=0.014), non-lobular histology (p=0.024), adjuvant chemotherapy (p=0.006), and various immune cell populations (tumor infiltrating lymphocytes (TILs), CD3+, CD8+, PD-1+ cells; all p<0.0001), PD-L1+ tumor cells (p<0.0001), and PD-L1+ stromal cells (p=0.003). Infiltration by TIGIT+ cells tended to be higher in non-molecular apocrine tumors (p=0.088). PVR was significantly associated with histological grade (p<0.0001), the basal-like (p=0.003) and non-molecular apocrine phenotypes (p=0.039), high TILs infiltration (p=0.011), CD3+ (p=0.002), CD8+ (p=0.024) T cells, and PD-L1 expression in tumor (p=0.003) and stromal cells (p=0.001). In univariate analysis, only known prognostic factors (age, tumor size, lymph node status, adjuvant chemotherapy, TILs and CD3+ T-cell infiltrate) were significantly associated with relapse-free survival (RFS) and overall survival. High TIGIT and PVR expression levels tended to be associated with longer RFS (p=0.079 and 0.045, respectively). The analysis that included only non-molecular apocrine TNBC revealed longer RFS for tumors that strongly expressed TIGIT or PVR (p=0.025 for TIGIT and 0.032 for PVR). Conclusions These results indicated that in TNBC, TIGIT+ cells can easily interact with PVR to exert their inhibitory effects. Their wide expression in TNBC and their association with other immune checkpoint components suggest the therapeutic interest of the TIGIT-PVR axis.
Collapse
Affiliation(s)
- Florence Boissière-Michot
- Translational Research Unit, Montpellier Cancer Institute Val d’Aurelle, Montpellier, France,*Correspondence: Florence Boissière-Michot,
| | - Marie-Christine Chateau
- Translational Research Unit, Montpellier Cancer Institute Val d’Aurelle, Montpellier, France
| | - Simon Thézenas
- Biometrics Unit, Montpellier Cancer Institute Val d’Aurelle, Montpellier, France
| | - Séverine Guiu
- Department of Medical Oncology, Montpellier Cancer Institute Val d’Aurelle, Montpellier, France,Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm U1194, Montpellier, France
| | - Angélique Bobrie
- Department of Medical Oncology, Montpellier Cancer Institute Val d’Aurelle, Montpellier, France,Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm U1194, Montpellier, France
| | - William Jacot
- Translational Research Unit, Montpellier Cancer Institute Val d’Aurelle, Montpellier, France,Department of Medical Oncology, Montpellier Cancer Institute Val d’Aurelle, Montpellier, France,Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm U1194, Montpellier, France,Faculty of Medicine, Montpellier University, Montpellier, France
| |
Collapse
|
12
|
Haack AM, Overall CM, Auf dem Keller U. Degradomics technologies in matrisome exploration. Matrix Biol 2022; 114:1-17. [PMID: 36280126 DOI: 10.1016/j.matbio.2022.10.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 10/05/2022] [Accepted: 10/20/2022] [Indexed: 11/06/2022]
Abstract
Consisting of a defined set of extracellular proteins secreted from resident cells and with minor contributions from serum proteins, the extracellular matrix (ECM) is an essential component of all tissues. Maintaining tissue homeostasis, structural support and cellular control through cell-ECM communication, the ECM has come to be viewed as not just a passive structural entity but rather as a dynamic signaling conduit between cells and the extracellular compartment. Proteins and their cleavage products mediate this communication, and aberrant signaling, either directly or indirectly distorting the ECM, results in pathological conditions including cancer, inflammation, fibrosis, and neurodegenerative diseases. Characterization of ECM components, the matrisome, the extracellular environment and their changes in disease is therefore of importance to understand and mitigate by developing novel therapeutics. Liquid chromatography-mass spectrometry (LC-MS) proteomics has been integral to protein and proteome research for decades and long superseded the obsolescent gel-based approaches. A continuous effort has ensured progress with increased sensitivity and throughput as more advanced equipment has been developed hand in hand with specialized enrichment, detection, and identification methods. Part of this effort lies in the field of degradomics, a branch of proteomics focused on discovering novel protease substrates by identification of protease-generated neo-N termini, the N-terminome, and characterizing the responsible protease networks. Various methods to do so have been developed, some specialized for specific tissue types, others for particular proteases, throughput, or ease of use. This review aims to provide an overview of the state-of-the-art proteomics techniques that have successfully been recently utilized to characterize proteolytic cleavages in the ECM and thereby guided new research and understanding of the ECM and matrisome biology.
Collapse
Affiliation(s)
- Aleksander M Haack
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads, DK-2800 Kongens Lyngby, Denmark
| | - Christopher M Overall
- Department of Biochemistry and Molecular Biology, Department of Oral Biological and Medical Sciences, Centre for Blood Research, University of British Columbia, 4.401 Life Sciences Institute, 2350 Health Sciences Mall, Vancouver, British Columbia V6T 1Z3, Canada.
| | - Ulrich Auf dem Keller
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Søltofts Plads, DK-2800 Kongens Lyngby, Denmark.
| |
Collapse
|
13
|
Xie J, Zheng S, Zou Y, Tang Y, Tian W, Wong CW, Wu S, Ou X, Zhao W, Cai M, Xie X. Turning up a new pattern: Identification of cancer-associated fibroblast-related clusters in TNBC. Front Immunol 2022; 13:1022147. [PMID: 36275659 PMCID: PMC9583405 DOI: 10.3389/fimmu.2022.1022147] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 09/16/2022] [Indexed: 11/17/2022] Open
Abstract
Growing evidence indicates a connection between cancer-associated fibroblasts (CAFs) and tumor microenvironment (TME) remodeling and tumor progression. Nevertheless, how patterns of CAFs impact TME and immunotherapy responsiveness in triple-negative breast cancer (TNBC) remains unclear. Here, we systematically investigate the relationship between TNBC progression and patterns of CAFs. By using unsupervised clustering methods in the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) dataset, we identified two distinct CAF-associated clusters that were related to clinical features, characteristics of TME, and prognosis of patients. Then, we established a CAF-related prognosis index (CPI) by the least absolute shrinkage and selection operator (LASSO)-Cox regression method. CPI showed prognostic accuracy in both training and validation cohorts (METABRIC, GSE96058, and GSE21653). Consequently, we constructed a nomogram with great predictive performance. Moreover, the CPI was verified to be correlated with the responsiveness of immunotherapy in three independent cohorts (GSE91061, GSE165252, and GSE173839). Taken together, the CPI might help us improve our recognition of the TME of TNBC, predict the prognosis of TNBC patients, and offer more immunotherapy strategies in the future.
Collapse
Affiliation(s)
- Jindong Xie
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Shaoquan Zheng
- Breast Disease Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yutian Zou
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yuhui Tang
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Wenwen Tian
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Chau-Wei Wong
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Song Wu
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Xueqi Ou
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Wanzhen Zhao
- Department of Radiotherapy, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Manbo Cai
- Department of Radiotherapy, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- *Correspondence: Xiaoming Xie, ; Manbo Cai,
| | - Xiaoming Xie
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- *Correspondence: Xiaoming Xie, ; Manbo Cai,
| |
Collapse
|
14
|
Mohindra R, Mohindra R, Agrawal DK, Thankam FG. Bioactive extracellular matrix fragments in tendon repair. Cell Tissue Res 2022; 390:131-140. [PMID: 36074173 DOI: 10.1007/s00441-022-03684-z] [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: 01/06/2022] [Accepted: 08/30/2022] [Indexed: 11/02/2022]
Abstract
Tendinopathy is a common tendon disorder that causes pain, loss of strength and function, and local inflammation mainly characterized by hypoxia, collagen degradation, and extracellular matrix (ECM) disorganization. Generally, ECM degradation and remodeling is tightly regulated; however, hyperactivation of matrix metalloproteases (MMPs) contributes to excessive collagenolysis under pathologic conditions resulting in tendon ECM degradation. This review article focuses on the production, function, and signaling of matrikines for tendon regeneration following injury with insights into the expression, tissue compliance, and cell proliferation exhibited by various matrikines. Furthermore, the regenerative properties suggest translational significance of matrikines to improve the outcomes post-injury by assisting with tendon healing.
Collapse
Affiliation(s)
- Ritika Mohindra
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766-1854, USA
| | - Rohit Mohindra
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766-1854, USA
| | - Devendra K Agrawal
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766-1854, USA
| | - Finosh G Thankam
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766-1854, USA.
| |
Collapse
|
15
|
Nishi K, Fu W, Kiyama R. Novel estrogen-responsive genes (ERGs) for the evaluation of estrogenic activity. PLoS One 2022; 17:e0273164. [PMID: 35976950 PMCID: PMC9385026 DOI: 10.1371/journal.pone.0273164] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 08/03/2022] [Indexed: 11/19/2022] Open
Abstract
Estrogen action is mediated by various genes, including estrogen-responsive genes (ERGs). ERGs have been used as reporter-genes and markers for gene expression. Gene expression profiling using a set of ERGs has been used to examine statistically reliable transcriptomic assays such as DNA microarray assays and RNA sequencing (RNA-seq). However, the quality of ERGs has not been extensively examined. Here, we obtained a set of 300 ERGs that were newly identified by six sets of RNA-seq data from estrogen-treated and control human breast cancer MCF-7 cells. The ERGs exhibited statistical stability, which was based on the coefficient of variation (CV) analysis, correlation analysis, and examination of the functional association with estrogen action using database searches. A set of the top 30 genes based on CV ranking were further evaluated quantitatively by RT-PCR and qualitatively by a functional analysis using the GO and KEGG databases and by a mechanistic analysis to classify ERα/β-dependent or ER-independent types of transcriptional regulation. The 30 ERGs were characterized according to (1) the enzymes, such as metabolic enzymes, proteases, and protein kinases, (2) the genes with specific cell functions, such as cell-signaling mediators, tumor-suppressors, and the roles in breast cancer, (3) the association with transcriptional regulation, and (4) estrogen-responsiveness. Therefore, the ERGs identified here represent various cell functions and cell signaling pathways, including estrogen signaling, and thus, may be useful to evaluate estrogenic activity.
Collapse
Affiliation(s)
- Kentaro Nishi
- Department of Life Science, Faculty of Life Science, Kyushu Sangyo University Matsukadai, Higashi-ku, Fukuoka, Japan
| | - Wenqiang Fu
- Department of Life Science, Faculty of Life Science, Kyushu Sangyo University Matsukadai, Higashi-ku, Fukuoka, Japan
| | - Ryoiti Kiyama
- Department of Life Science, Faculty of Life Science, Kyushu Sangyo University Matsukadai, Higashi-ku, Fukuoka, Japan
| |
Collapse
|
16
|
Binay S, Kaptan E. Transcription factor Runx2 changes the expression of some matricellular proteins in metastatic breast cancer cells. Mol Biol Rep 2022; 49:6433-6441. [PMID: 35441354 DOI: 10.1007/s11033-022-07457-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 04/05/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Runx2 is one of the runt-related genes that are overexpressed in human cancers and contribute to metastasis. The cancer cell metastasis requires modifications of the extracellular matrix (ECM) and reduction in ECM-cell interaction. This process is performed by various enzymes and proteins secreted by cancer and surrounding cells. This study aimed to investigate the effect of the Runx2 transcription factor on the expression of matricellular proteins such as HPA1, LOX, SPARC, and OPN, which have important roles in ECM modification and ECM-cell interaction in human breast cancer. Also, the changes in their associated oncogenic pathways including Akt, Erk, FAK activities, and c-jun protein expression were investigated. METHODS AND RESULTS Runx2 knockdown model was created using runx2 siRNA in MDA-MB-231 human metastatic breast cancer cells. The changes in the mRNA and protein expressions of ECM proteins were shown by the qPCR and Western blotting, respectively. The results showed that there was a decrease in both mRNA and protein expressions of HPA1, SPARC, and LOX, whereas there was no change in those of OPN. Phosphorylated Akt, Erk, FAK levels, and protein expression of c-jun, however, decreased in the cells. CONCLUSION Our results revealed that Runx2 affected matricellular protein expression, which is important for metastasis and invasion of breast cancer. Hence, we have concluded that runx2 appears to be efficient for regulating breast cancer metastasis through an expression of matricellular proteins.
Collapse
Affiliation(s)
- Sevgi Binay
- Faculty of Science, Department of Biology, Istanbul University, Vezneciler, 34134, Istanbul, Turkey
| | - Engin Kaptan
- Faculty of Science, Department of Biology, Istanbul University, Vezneciler, 34134, Istanbul, Turkey.
| |
Collapse
|
17
|
Deng S, Zhang L, Li J, Jin Y, Wang J. Activation of the PI3K-AKT signaling pathway by SPARC contributes to the malignant phenotype of cholangiocarcinoma cells. Tissue Cell 2022; 76:101756. [PMID: 35217388 DOI: 10.1016/j.tice.2022.101756] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 02/07/2022] [Accepted: 02/09/2022] [Indexed: 12/22/2022]
Abstract
Cholangiocarcinoma (CCA) is a primary biliary epithelium malignancy with limited therapies, poor prognosis and high mortality rate. Nowadays, the molecular mechanisms of CCA remain elusive. SPARC has been proposed to be highly expressed in clinical CCA tissues, but few studies has been elucidated its functions in CCA. In the current study, we aimed to investigate the functional role of SPARC in the progression of CCA. In this study, a significantly increased expression of SPARC was observed in CCA tissues and cells. Knockdown of SPARC by RNA interference significantly impeded the proliferation of CCA cells. Moreover, SPARC silencing hampered the migration and invasion of CCA cells by inhibiting EMT. In parallel, overexpression of SPARC in RBE cells had the opposite effects. Mechanically, SPARC promoted proliferation, migration, invasion, and EMT of CCA cells in vitro via activating the PI3K-AKT signaling. Overall, our integrated analysis revealed that SPARC plays a crucial role in CCA progression via the PI3K-AKT signaling pathway, which suggests that targeting SPARC might represent a promising approach for improving CCA patient's clinical outcome.
Collapse
Affiliation(s)
- Shikang Deng
- Medical School, Kunming University of Science and Technology, Kumming, Yunnan, China; Department of Hepatobiliary and Pancreatic Surgery, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kumming, Yunnan, China
| | - Li Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kumming, Yunnan, China
| | - Jiao Li
- Department of Hepatobiliary and Pancreatic Surgery, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kumming, Yunnan, China
| | - Yan Jin
- Department of Hepatobiliary and Pancreatic Surgery, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kumming, Yunnan, China
| | - Junfeng Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First People's Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kumming, Yunnan, China.
| |
Collapse
|
18
|
Seo SU, Woo SM, Im SS, Jang Y, Han E, Kim SH, Lee H, Lee HS, Nam JO, Gabrielson E, Min KJ, Kwon TK. Cathepsin D as a potential therapeutic target to enhance anticancer drug-induced apoptosis via RNF183-mediated destabilization of Bcl-xL in cancer cells. Cell Death Dis 2022; 13:115. [PMID: 35121737 PMCID: PMC8816936 DOI: 10.1038/s41419-022-04581-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 01/11/2022] [Accepted: 01/26/2022] [Indexed: 12/17/2022]
Abstract
Cathepsin D (Cat D) is well known for its roles in metastasis, angiogenesis, proliferation, and carcinogenesis in cancer. Despite Cat D being a promising target in cancer cells, effects and underlying mechanism of its inhibition remain unclear. Here, we investigated the plausibility of using Cat D inhibition as an adjuvant or sensitizer for enhancing anticancer drug-induced apoptosis. Inhibition of Cat D markedly enhanced anticancer drug-induced apoptosis in human carcinoma cell lines and xenograft models. The inhibition destabilized Bcl-xL through upregulation of the expression of RNF183, an E3 ligase of Bcl-xL, via NF-κB activation. Furthermore, Cat D inhibition increased the proteasome activity, which is another important factor in the degradation of proteins. Cat D inhibition resulted in p62-dependent activation of Nrf2, which increased the expression of proteasome subunits (PSMA5 and PSMB5), and thereby, the proteasome activity. Overall, Cat D inhibition sensitized cancer cells to anticancer drugs through the destabilization of Bcl-xL. Furthermore, human renal clear carcinoma (RCC) tissues revealed a positive correlation between Cat D and Bcl-xL expression, whereas RNF183 and Bcl-xL expression indicated inverse correlation. Our results suggest that inhibition of Cat D is promising as an adjuvant or sensitizer for enhancing anticancer drug-induced apoptosis in cancer cells.
Collapse
|
19
|
Zheng S, Zou Y, Tang Y, Yang A, Liang JY, Wu L, Tian W, Xiao W, Xie X, Yang L, Xie J, Wei W, Xie X. Landscape of cancer-associated fibroblasts identifies the secreted biglycan as a protumor and immunosuppressive factor in triple-negative breast cancer. Oncoimmunology 2022; 11:2020984. [PMID: 35003899 PMCID: PMC8741292 DOI: 10.1080/2162402x.2021.2020984] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cancer-associated fibroblasts (CAFs) are essential for tumor microenvironment remodeling and correlate with tumor progression. However, interactions between CAFs and tumor cells and immune cells in triple-negative breast cancer (TNBC) are still poorly explored. Here, we investigate the role of CAFs in TNBC and potential novel mediators of their functions. The clustering of classic markers was applied to estimate the relative abundance of CAFs in TNBC cohorts. Primary fibroblasts were isolated from normal and tumor samples. The RNA and culture medium of fibroblasts were subjected to RNA sequencing and mass spectrometry to explore the upregulated signatures in CAFs. Microdissection and single-cell RNA sequencing datasets were used to examine the expression profiles. CAFs were associated with hallmark signalings and immune components in TNBC. Clustering based on CAF markers in the literature revealed different CAF infiltration groups in TNBC: low, medium and high. Most of the cancer hallmark signaling pathways were enriched in the high CAF infiltration group. Furthermore, RNA sequencing and mass spectrometry identified biglycan (BGN), a soluble secreted protein, as upregulated in CAFs compared to normal cancer-adjacent fibroblasts (NAFs). The expression of biglycan was negatively correlated with CD8 + T cells. Biglycan indicated poor prognostic outcomes and might be correlated with the immunosuppressive tumor microenvironment (TME). In conclusion, CAFs play an essential role in tumor progression and the TME. We identified an extracellular protein, biglycan, as a prognostic marker and potential therapeutic target in TNBC.
Collapse
Affiliation(s)
- Shaoquan Zheng
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Yutian Zou
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Yuhui Tang
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Anli Yang
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Jie-Ying Liang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China.,Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Linyu Wu
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Wenwen Tian
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Weikai Xiao
- Department of Breast Cancer, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Xinhua Xie
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Lu Yang
- Department of Radiotherapy, Cancer Center, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Jindong Xie
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Weidong Wei
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Xiaoming Xie
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| |
Collapse
|
20
|
SPARC-mediated long-term retention of nab-paclitaxel in pediatric sarcomas. J Control Release 2021; 342:81-92. [PMID: 34974029 DOI: 10.1016/j.jconrel.2021.12.035] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 12/24/2021] [Accepted: 12/28/2021] [Indexed: 12/29/2022]
Abstract
Secreted protein acidic and rich in cysteine (SPARC) is a matricellular glycoprotein overexpressed by several cancers. Because SPARC shows high binding affinity to albumin, we reasoned that pediatric sarcoma xenografts expressing SPARC would show enhanced uptake and accumulation of nanoparticle albumin-bound (nab)-paclitaxel, a potent anticancer drug formulation. We first evaluated the expression of SPARC in patient-derived xenografts (PDXs) of Ewing sarcoma, rhabdomyosarcoma and osteosarcoma, finding variable SPARC gene expression that correlated well with SPARC protein measured by immunoblotting. We revealed that the activity of the fusion gene chimera EWSR1-FLI1, the genetic driver of Ewing sarcoma, leads to lower expression of the gene SPARC in these tumors, likely due to enriched acetylation marks of the histone H3 lysine 27 at regions including the SPARC promoter and potential enhancers. Then, we used SPARC-edited Ewing sarcoma cells (A673 line) to demonstrate that SPARC knocked down (KD) cells accumulated significantly less amount of nab-paclitaxel in vitro than SPARC wild type (WT) cells. In vivo, SPARC KD and SPARC WT subcutaneous xenografts in mice achieved similar maximum intratumoral concentrations of nab-paclitaxel, though drug clearance from SPARC WT tumors was significantly slower. We confirmed such SPARC-mediated long-term intratumoral accumulation of nab-paclitaxel in Ewing sarcoma PDX with high expression of SPARC, which accumulated significantly more nab-paclitaxel than SPARC-low PDX. SPARC-high PDX responded better to nab-paclitaxel than SPARC-low tumors, although these results should be taken cautiously, given that the PDXs were established from different patients that could have specific determinants predisposing response to paclitaxel. In addition, SPARC KD Ewing sarcoma xenografts responded better to soluble docetaxel and paclitaxel than to nab-paclitaxel, while SPARC WT ones showed similar response to soluble and albumin-carried drugs. Overall, our results show that pediatric sarcomas expressing SPARC accumulate nab-paclitaxel for longer periods of time, which could have clinical implications for chemotherapy efficacy.
Collapse
|
21
|
Liao J, Zheng Y, Hu M, Xu P, Lin L, Liu X, Wu Y, Huang B, Ye X, Li S, Duan R, Fu H, Huang J, Wen L, Fu Y, Kilby MD, Kenny LC, Baker PN, Qi H, Tong C. Impaired Sphingosine-1-Phosphate Synthesis Induces Preeclampsia by Deactivating Trophoblastic YAP (Yes-Associated Protein) Through S1PR2 (Sphingosine-1-Phosphate Receptor-2)-Induced Actin Polymerizations. Hypertension 2021; 79:399-412. [PMID: 34865521 DOI: 10.1161/hypertensionaha.121.18363] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Incomplete spiral artery remodeling, caused by impaired extravillous trophoblast invasion, is a fundamental pathogenic process associated with malplacentation and the development of preeclampsia. Nevertheless, the mechanisms controlling this regulation of trophoblast invasion are largely unknown. We report that sphingosine-1-phosphate synthesis and expression is abundant in healthy trophoblast, whereas in pregnancies complicated by preeclampsia the placentae are associated with reduced sphingosine-1-phosphate and lower SPHK1 (sphingosine kinase 1) expression and activity. In vivo inhibition of sphingosine kinase 1 activity during placentation in pregnant mice led to decreased placental sphingosine-1-phosphate production and defective placentation, resulting in a preeclampsia phenotype. Moreover, sphingosine-1-phosphate increased HTR8/SVneo (immortalized trophoblast cells) cell invasion in a Hippo-signaling-dependent transcriptional coactivator YAP (Yes-associated protein) dependent manner, which is activated by S1PR2 (sphingosine-1-phosphate receptor-2) and downstream RhoA/ROCK induced actin polymerization. Mutation-based YAP-5SA demonstrated that sphingosine-1-phosphate activation of YAP could be either dependent or independent of Hippo signaling. Together, these findings suggest a novel pathogenic pathway of preeclampsia via disrupted sphingosine-1-phosphate metabolism and signaling-induced, interrupted actin dynamics and YAP deactivation; this may lead to potential novel intervention targets for the prevention and management of preeclampsia.
Collapse
Affiliation(s)
- Jiujiang Liao
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Ministry of Education-International Collaborative Laboratory of Reproduction and Development, Chongqing, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.)
| | - Yangxi Zheng
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Department of Biochemistry & Molecular Biology, University of Texas McGovern Medical School at Houston (Y.Z.).,Department of Stem Cell Transplantation and Cell Therapy, MD Anderson Cancer Center, Houston, TX (Y.Z.)
| | - Mingyu Hu
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Ministry of Education-International Collaborative Laboratory of Reproduction and Development, Chongqing, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.)
| | - Ping Xu
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Ministry of Education-International Collaborative Laboratory of Reproduction and Development, Chongqing, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.)
| | - Li Lin
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Ministry of Education-International Collaborative Laboratory of Reproduction and Development, Chongqing, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.)
| | - Xiyao Liu
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Ministry of Education-International Collaborative Laboratory of Reproduction and Development, Chongqing, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.)
| | - Yue Wu
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Ministry of Education-International Collaborative Laboratory of Reproduction and Development, Chongqing, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.)
| | - Biao Huang
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Ministry of Education-International Collaborative Laboratory of Reproduction and Development, Chongqing, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.)
| | - Xuan Ye
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Ministry of Education-International Collaborative Laboratory of Reproduction and Development, Chongqing, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.)
| | - Sisi Li
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Ministry of Education-International Collaborative Laboratory of Reproduction and Development, Chongqing, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.)
| | - Ran Duan
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Ministry of Education-International Collaborative Laboratory of Reproduction and Development, Chongqing, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.)
| | - Huijia Fu
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Ministry of Education-International Collaborative Laboratory of Reproduction and Development, Chongqing, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.)
| | - Jiayu Huang
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Ministry of Education-International Collaborative Laboratory of Reproduction and Development, Chongqing, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.)
| | - Li Wen
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Ministry of Education-International Collaborative Laboratory of Reproduction and Development, Chongqing, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.)
| | - Yong Fu
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Ministry of Education-International Collaborative Laboratory of Reproduction and Development, Chongqing, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.)
| | - Mark D Kilby
- Institute of Metabolism and System Research, College of Medical & Dental Sciences, University of Birmingham and the Fetal Medicine Centre, Birmingham Women's and Children's Foundation Trust, United Kingdom (M.D.K.)
| | - Louise C Kenny
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, Faculty of Health and Life Sciences, University of Liverpool, United Kingdom (L.C.K.)
| | - Philip N Baker
- College of Life Sciences, University of Leicester, United Kingdom (P.N.B.)
| | - Hongbo Qi
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Ministry of Education-International Collaborative Laboratory of Reproduction and Development, Chongqing, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Chongqing Women and Children's Health Center, China (H.Q.)
| | - Chao Tong
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, China (J.L., Y.Z., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,Ministry of Education-International Collaborative Laboratory of Reproduction and Development, Chongqing, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.).,State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, China (J.L., M.H., P.X., L.L., X.L., Y.W., B.H., X.Y., S.L., R.D., H.F., J.H., L.W., Y.F., H.Q., C.T.)
| |
Collapse
|
22
|
Mintoo M, Chakravarty A, Tilvawala R. N-Terminomics Strategies for Protease Substrates Profiling. Molecules 2021; 26:molecules26154699. [PMID: 34361849 PMCID: PMC8348681 DOI: 10.3390/molecules26154699] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/20/2021] [Accepted: 07/22/2021] [Indexed: 01/02/2023] Open
Abstract
Proteases play a central role in various biochemical pathways catalyzing and regulating key biological events. Proteases catalyze an irreversible post-translational modification called proteolysis by hydrolyzing peptide bonds in proteins. Given the destructive potential of proteolysis, protease activity is tightly regulated. Dysregulation of protease activity has been reported in numerous disease conditions, including cancers, neurodegenerative diseases, inflammatory conditions, cardiovascular diseases, and viral infections. The proteolytic profile of a cell, tissue, or organ is governed by protease activation, activity, and substrate specificity. Thus, identifying protease substrates and proteolytic events under physiological conditions can provide crucial information about how the change in protease regulation can alter the cellular proteolytic landscape. In recent years, mass spectrometry-based techniques called N-terminomics have become instrumental in identifying protease substrates from complex biological mixtures. N-terminomics employs the labeling and enrichment of native and neo-N-termini peptides, generated upon proteolysis followed by mass spectrometry analysis allowing protease substrate profiling directly from biological samples. In this review, we provide a brief overview of N-terminomics techniques, focusing on their strengths, weaknesses, limitations, and providing specific examples where they were successfully employed to identify protease substrates in vivo and under physiological conditions. In addition, we explore the current trends in the protease field and the potential for future developments.
Collapse
|