1
|
Lu X, Friedrich LJ, Efferth T. Natural products targeting tumour angiogenesis. Br J Pharmacol 2025; 182:2094-2136. [PMID: 37680009 DOI: 10.1111/bph.16232] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/15/2023] [Accepted: 08/28/2023] [Indexed: 09/09/2023] Open
Abstract
Tumour angiogenesis is the formation of new blood vessels to support the growth of a tumour. This process is critical for tumour progression and metastasis, making it an attractive approach to cancer therapy. Natural products derived from plants, animals or microorganisms exert anti-angiogenic properties and can be used to inhibit tumour growth and progression. In this review, we comprehensively report on the current status of natural products against tumour angiogenesis from four perspectives until March 2023: (1) the role of pro-angiogenic factors and antiangiogenic factors in tumour angiogenesis; (2) the development of anti-tumour angiogenesis therapy (monoclonal antibodies, VEGFR-targeted small molecules and fusion proteins); (3) the summary of anti-angiogenic natural agents, including polyphenols, polysaccharides, alkaloids, terpenoids, saponins and their mechanisms of action, and (4) the future perspectives of anti-angiogenic natural products (bioavailability improvement, testing of dosage and side effects, combination use and discovery of unique natural-based compounds). Our review aims to better understand the potential of natural products for drug development in inhibiting tumour angiogenesis and further aid the effective transition of these outcomes into clinical trials. LINKED ARTICLES: This article is part of a themed issue Natural Products and Cancer: From Drug Discovery to Prevention and Therapy. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v182.10/issuetoc.
Collapse
Affiliation(s)
- Xiaohua Lu
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Lara Johanna Friedrich
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
2
|
Guo J, Bai R, Luo R, Lin L, Zheng Y. Angiostatin: a promising therapeutic target for atopic dermatitis. Arch Dermatol Res 2025; 317:616. [PMID: 40119948 DOI: 10.1007/s00403-025-04126-w] [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: 10/05/2024] [Revised: 02/14/2025] [Accepted: 02/25/2025] [Indexed: 03/25/2025]
Abstract
Angiostatin, a 38-45 KDa proteolytic fragment derived from plasminogen, has garnered significant attention for its dual roles in inhibiting angiogenesis and modulating inflammation. We employed bidirectional Mendelian randomization (MR), meta-analysis, and colocalization to investigate the causal relationship between angiostatin and atopic dermatitis (AD) using three angiostatin and two AD datasets. Additionally, we analyzed global epidemiological trends (1990-2021) and performed transcriptomic profiling of AD. MR analyses revealed a protective effect of angiostatin on AD risk (combined odds ratio: 0.9437, 95% confidence interval [CI]: 0.9198-0.9683, p < 0.0001), while reverse analyses showed no association (standardized mean difference: -0.0029, 95% CI: -0.0516-0.0459, p = 0.9084). Colocalization indicated no shared causal variants (H4 probabilities < 80%). Epidemiological trends highlighted declining age-standardized AD rates despite rising case numbers. Transcriptomic analyses implicated NF-κB, PI3K-Akt, and JAK-STAT pathways in AD pathogenesis. These findings position angiostatin as a dual-action therapeutic candidate, offering novel opportunities to simultaneously target vascular remodeling and immune dysregulation in AD. Translational research is warranted to harness its clinical potential.
Collapse
Affiliation(s)
- Jiaqi Guo
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Ruimin Bai
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Ruiting Luo
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Liyan Lin
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yan Zheng
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.
| |
Collapse
|
3
|
Peeney D, Kumar S, Singh TP, Liu Y, Jensen SM, Chowdhury A, Coates-Park S, Rich J, Gurung S, Fan Y, Meerzaman D, Stetler-Stevenson WG. Timp2 loss-of-function mutation and TIMP2 treatment in a murine model of NSCLC: Modulation of immunosuppression and oncogenic signaling. Transl Oncol 2025; 53:102309. [PMID: 39904284 PMCID: PMC11846589 DOI: 10.1016/j.tranon.2025.102309] [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: 07/31/2024] [Revised: 12/06/2024] [Accepted: 01/30/2025] [Indexed: 02/06/2025] Open
Abstract
Mounting evidence suggests that the tissue inhibitor of metalloproteinases-2 (TIMP2) can reduce tumor burden and metastasis. However, the demonstration of such anti-tumor activity and associated mechanisms using in vivo tumor models is lacking. The effects of a Timp2 functional mutation and administration of recombinant TIMP2 were examined in both orthotopic and heterotopic murine models of lung cancer using C57Bl/6 syngeneic Lewis Lung 2-luciferase 2 cells (LL2-Luc2) cells. Mice harboring a functional mutation of TIMP2 (mT2) display markedly increased primary lung tumor growth, increased mortality, enriched vasculature, and enhanced infiltration of pro-tumorigenic, immunosuppressive myeloid cells. Treatment with recombinant TIMP2 reduced primary tumor growth in both mutant and wild-type (wt) mice. Comparison of transcriptional profiles of lung tissues from tumor-free, wt versus mT2 mice reveals only minor changes. However, lung tumor-bearing mice of both genotypes demonstrate significant genotype-dependent changes in gene expression following treatment with TIMP. In tumor-bearing wt mice, TIMP2 treatment reduced the expression of upstream oncogenic mediators, whereas treatment of mT2 mice resulted in an immunomodulatory phenotype. A heterotopic subcutaneous model generating metastatic pulmonary tumors demonstrated that daily administration of recombinant TIMP2 significantly reduces the expression of heat shock proteins, suggesting a reduction of cell-stress responses. In summary, we describe how TIMP2 exerts novel, anti-tumor effects in a murine model of lung cancer and that rTIMP2 treatment supports a normalizing effect on the tumor microenvironment. Our findings show that TIMP2 treatment demonstrates significant potential as an adjuvant in the treatment of NSCLC.
Collapse
Affiliation(s)
- David Peeney
- Extracellular Matrix Pathology Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute (NCI), Bethesda, MD 20892, USA.
| | - Sarvesh Kumar
- Extracellular Matrix Pathology Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute (NCI), Bethesda, MD 20892, USA
| | - Tej Pratap Singh
- Laboratory of Molecular Immunology, National Institute for Allergy, and Infectious Disease (NIAID), Bethesda, MD 20892, USA
| | - Yueqin Liu
- Extracellular Matrix Pathology Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute (NCI), Bethesda, MD 20892, USA
| | - Sandra M Jensen
- Extracellular Matrix Pathology Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute (NCI), Bethesda, MD 20892, USA
| | - Ananda Chowdhury
- Extracellular Matrix Pathology Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute (NCI), Bethesda, MD 20892, USA
| | - Sasha Coates-Park
- Extracellular Matrix Pathology Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute (NCI), Bethesda, MD 20892, USA
| | - Joshua Rich
- Extracellular Matrix Pathology Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute (NCI), Bethesda, MD 20892, USA
| | - Sadeechya Gurung
- Extracellular Matrix Pathology Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute (NCI), Bethesda, MD 20892, USA
| | - Yu Fan
- Computational Genomics and Bioinformatics Group, Center for Biomedical Informatics & Information Technology, National Cancer Institute, Rockville, MD 20850, USA
| | - Daoud Meerzaman
- Computational Genomics and Bioinformatics Group, Center for Biomedical Informatics & Information Technology, National Cancer Institute, Rockville, MD 20850, USA
| | - William G Stetler-Stevenson
- Extracellular Matrix Pathology Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute (NCI), Bethesda, MD 20892, USA.
| |
Collapse
|
4
|
Chen L, Liao K, Zhang Y, Zheng S, He J, Tang H, Wu H, Zhong W, Li S, Li Y. Association of GWAS-Reported Variant of Matrix Metalloproteinase 12 Gene with Susceptibility to Ischemic Stroke in Southern Chinese Population. J Inflamm Res 2024; 17:9231-9241. [PMID: 39583862 PMCID: PMC11585993 DOI: 10.2147/jir.s487321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 11/14/2024] [Indexed: 11/26/2024] Open
Abstract
Background Accumulating evidence suggests that matrix metalloproteinase (MMP) 12 plays a detrimental role in cerebro-cardiovascular diseases, including ischemic stroke (IS). Previous genome-wide association studies (GWAS) correlated the MMP12 rs660599 variant to IS risk in Europeans. However, this association is yet to be elucidated in the Chinese population. This study aims to assess the genetic predisposition of the MMP12 rs660599 G > A variant with regard to IS risk and short-term outcomes in individuals from Southern China. Methods The Multiplex SNaPshot assay was used to genotype rs660599 in 1035 IS patients and 1061 age-matched healthy controls. Multivariate logistic regression analyses evaluated the effect of the rs660599 G > A polymorphism on IS susceptibility and short-term outcomes. Results No significant association was found between the rs660599 G > A polymorphism and IS risk, even in dominant and recessive models. However, a relationship between rs660599 genotypes and diabetic status revealed that carriers of the A allele and the GA/AA genotype were more likely to develop IS. The presence of diabetes exacerbated the larger infarct volumes and elevated serum MMP12 levels seen in IS patients with the rs660599 A allele. The A allele of rs660599 and the GA/AA genotype were both correlated to moderate and severe stroke with poor short-term outcomes. Conclusion The MMP12 rs660599 polymorphism is associated with a higher incidence of IS in people with diabetes and can serve as a biomarker for assessing the severity of IS and its short-term consequences.
Collapse
Affiliation(s)
- Linfa Chen
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, People’s Republic of China
- Department of Neurology, Huizhou Third People’s Hospital, Guangzhou Medical University, Huizhou, People’s Republic of China
| | - Keqi Liao
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, People’s Republic of China
- Institute of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, People’s Republic of China
| | - Yutian Zhang
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, People’s Republic of China
- Institute of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, People’s Republic of China
| | - Shutao Zheng
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, People’s Republic of China
- Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, People’s Republic of China
| | - Jiawen He
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, People’s Republic of China
- Institute of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, People’s Republic of China
| | - Henglei Tang
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, People’s Republic of China
- Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, People’s Republic of China
| | - Hailing Wu
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, People’s Republic of China
- Institute of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, People’s Republic of China
| | - Wangtao Zhong
- Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, People’s Republic of China
| | - Shengnan Li
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, People’s Republic of China
- Institute of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, People’s Republic of China
| | - You Li
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, People’s Republic of China
- Institute of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, People’s Republic of China
| |
Collapse
|
5
|
Sharma D, Thomas S, Moody TB, Taylor M, Ituarte B, Georgeson CJ, Barrett CD, Wei EX. Laboratory and clinical haemostatic aberrations in primary dermatologic disease: A review. Thromb J 2024; 22:101. [PMID: 39533305 PMCID: PMC11558853 DOI: 10.1186/s12959-024-00665-w] [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: 07/09/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Inflammatory dermatologic diseases have long been viewed as a "skin limited" disease process. Current literature on inflammatory dermatologic diseases investigates their relationship and influence on thromboembolic states and thromboembolic complications and the understanding of their pathophysiology and molecular mechanisms.Studies specifically discuss known inflammatory skin diseases including alopecia areata, vitiligo, psoriasis, hidradenitis suppurativa, atopic dermatitis, chronic spontaneous urticaria, and autoimmune bullous diseases, and their effects on systemic inflammation, associated cardiovascular comorbidities, and thromboembolic or hypercoagulable states. The limited current literature shows potential for links between inflammatory skin diseases and hypercoagulable states. Biomarkers such as F1 + 2, D-dimer, eosinophilic cationic protein, and PAI-1 are currently being studied to outline the mechanisms connecting inflammatory skin disease to the coagulation system. Further study and larger amounts of data are needed to draw definitive conclusions, especially when interpreting biomarkers alone such as PAI-1.The mechanisms, rates of systemic inflammation, and clinical outcomes of traditionally "skin limited" inflammatory diseases remain chronically understudied in dermatology. Many organ systems have well established connections between inflammatory disease and hypercoagulable states, but there are significant gaps in the literature regarding skin diseases. There is a significant need for comprehensive investigation of molecular mechanisms behind inflammatory dermatologic disease and hypercoagulability, how hypercoagulability effects clinical outcomes, and proper intervention to optimize patient outcomes.
Collapse
Affiliation(s)
- Divya Sharma
- Department of Dermatology, University of Nebraska Medical Center, 985645, Omaha, NE, 68198-5645, USA
| | - Sierra Thomas
- Department of Dermatology, University of Nebraska Medical Center, 985645, Omaha, NE, 68198-5645, USA
- University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Trace B Moody
- Division of Acute Care Surgery, Department of Surgery, University of Nebraska Medical Center, Omaha, NE, USA
| | - Mitchell Taylor
- Department of Dermatology, University of Nebraska Medical Center, 985645, Omaha, NE, 68198-5645, USA
- Creighton University School of Medicine, Omaha, NE, USA
| | - Bianca Ituarte
- Department of Dermatology, University of Nebraska Medical Center, 985645, Omaha, NE, 68198-5645, USA
- University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA
| | - Corey J Georgeson
- Department of Dermatology, University of Nebraska Medical Center, 985645, Omaha, NE, 68198-5645, USA
| | - Christopher D Barrett
- Division of Acute Care Surgery, Department of Surgery, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Erin X Wei
- Department of Dermatology, University of Nebraska Medical Center, 985645, Omaha, NE, 68198-5645, USA
| |
Collapse
|
6
|
Shimizu K, Nishimura N, Wang T, Yamamoto T, Suzuki E, Hasumi K. Anti-angiogenic activity of a novel angiostatin-like plasminogen fragment produced by a bacterial metalloproteinase. Heliyon 2024; 10:e35232. [PMID: 39170245 PMCID: PMC11336434 DOI: 10.1016/j.heliyon.2024.e35232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 07/24/2024] [Accepted: 07/24/2024] [Indexed: 08/23/2024] Open
Abstract
Tumor growth depends on angiogenesis, a process by which new blood vessel are formed from pre-existing normal blood vessels. Proteolytic fragments of plasminogen, containing varying numbers of plasminogen kringle domains, collectively known as angiostatin, are a naturally occurring inhibitor of angiogenesis and inhibit tumor growth. We have developed an "affinity-capture reactor" that enables a single-step method for the production/purification of an angiostatin-like plasminogen fragment from human plasma using an immobilized bacterial metalloproteinase. The resulting fragment, named BL-angiostatin, contains one or two glycosyl chains and the N-terminal PAN module, which are not present in canonical angiostatins tested for cancer treatment. BL-angiostatin inhibited angiogenesis in vitro at 20 nM and the growth of both allograft and human xenograft tumors as well as lung metastasis of primary tumors mice at 0.3-10 mg kg-1. Derivatives of BL angiostatin lacking the PAN module or the terminal sialic acids in the glycosyl chains showed reduced anti-angiogenic activity in vivo, suggesting a role for these functions in activity, possibly via conferring a pharmacokinetic advantage to BL angiostatin compared to recombinant angiostatin lacking both features. These results highlight the potential of BL-angiostatin for therapeutic applications.
Collapse
Affiliation(s)
- Kosuke Shimizu
- Department of Applied Biological Science, Tokyo University of Agriculture and Technology, 3-5-8 Saiwaicho, Fuchu, Tokyo, 183-8509, Japan
- Division of Research and Development, TMS Co., Ltd., 1-9-11F Fuchucho, Fuchu, Tokyo, 183-0055, Japan
| | - Naoko Nishimura
- Division of Research and Development, TMS Co., Ltd., 1-9-11F Fuchucho, Fuchu, Tokyo, 183-0055, Japan
| | - Taolin Wang
- Department of Applied Biological Science, Tokyo University of Agriculture and Technology, 3-5-8 Saiwaicho, Fuchu, Tokyo, 183-8509, Japan
| | - Tetsuro Yamamoto
- Research Center, EPS Innovative Medicine, Kagurazaka AK Building, 1-8 Tsukudo-cho, Shinjuku-ku, Tokyo, 162-0821, Japan
| | - Eriko Suzuki
- Department of Applied Biological Science, Tokyo University of Agriculture and Technology, 3-5-8 Saiwaicho, Fuchu, Tokyo, 183-8509, Japan
| | - Keiji Hasumi
- Department of Applied Biological Science, Tokyo University of Agriculture and Technology, 3-5-8 Saiwaicho, Fuchu, Tokyo, 183-8509, Japan
- Division of Research and Development, TMS Co., Ltd., 1-9-11F Fuchucho, Fuchu, Tokyo, 183-0055, Japan
| |
Collapse
|
7
|
Veeravalli KK. Implications of MMP-12 in the pathophysiology of ischaemic stroke. Stroke Vasc Neurol 2024; 9:97-107. [PMID: 37336584 PMCID: PMC11103161 DOI: 10.1136/svn-2023-002363] [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: 02/03/2023] [Accepted: 06/05/2023] [Indexed: 06/21/2023] Open
Abstract
This article focuses on the emerging role of matrix metalloproteinase-12 (MMP-12) in ischaemic stroke (IS). MMP-12 expression in the brain increases dramatically in animal models of IS, and its suppression reduces brain damage and promotes neurological, sensorimotor and cognitive functional outcomes. Thus, MMP-12 could represent a potential target for the management of IS. This article provides an overview of MMP-12 upregulation in the brain following IS, its deleterious role in the post-stroke pathogenesis (blood-brain barrier disruption, inflammation, apoptosis and demyelination), possible molecular interactions and mechanistic insights, its involvement in post-ischaemic functional deficits and recovery as well as the limitations, perspectives, challenges and future directions for further research. Prior to testing any MMP-12-targeted therapy in patients with acute IS, additional research is needed to establish the effectiveness of MMP-12 suppression against IS in older animals and in animals with comorbidities. This article also examines the clinical implications of suppressing MMP-12 alone or in combination with MMP-9 for extending the currently limited tissue plasminogen activator therapy time window. Targeting of MMP-12 is expected to have a profound influence on the therapeutic management of IS in the future.
Collapse
Affiliation(s)
- Krishna Kumar Veeravalli
- Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, Illinois, USA
| |
Collapse
|
8
|
Dougan M, Nguyen LH, Buchbinder EI, Lazarus HM. Sargramostim for Prophylactic Management of Gastrointestinal Immune-Related Adverse Events of Immune Checkpoint Inhibitor Therapy for Cancer. Cancers (Basel) 2024; 16:501. [PMID: 38339253 PMCID: PMC10854719 DOI: 10.3390/cancers16030501] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/22/2024] [Accepted: 01/22/2024] [Indexed: 02/12/2024] Open
Abstract
Immune checkpoint inhibitor (ICI) therapy improves outcomes in several cancers. Unfortunately, many patients experience grade 3-4 treatment-related adverse events, including gastrointestinal (GI) toxicities which are common. These GI immune-related adverse events (irAEs) induced by ICIs present significant clinical challenges, require prompt intervention, and result in treatment delays or discontinuations. The treatment for these potentially severe and even fatal GI irAEs which include enterocolitis, severe diarrhea, and hepatitis may interfere with the anti-cancer approach. Sargramostim (glycosylated, yeast-derived, recombinant human GM-CSF) is an agent that has been used in clinical practice for more than 30 years with a well-recognized safety profile and has been studied in many therapeutic areas. The mechanism of action of sargramostim may treat moderate-to-severe GI irAEs without impairing the anti-cancer therapy. Some early data also suggest a potential survival benefit. Through the differentiation/maturation of monocytes, macrophages, and neutrophils and induction of anti-inflammatory T cell responses, GM-CSF aids in GI homeostasis, mucosal healing, and mucosal immunity. GM-CSF knockout mice are susceptible to severe colitis which was prevented with murine GM-CSF administration. For some patients with GI mucosa and immune cell function impairment, e.g., Crohn's disease, sargramostim reduces disease severity. In a prospective, randomized study (ECOG 1608), advanced melanoma patients had a reduction in grade 3-5 GI irAEs and less frequent colonic perforation in the sargramostim plus ipilimumab arm compared to ipilimumab alone. Sargramostim continues to be studied with ICIs for the prophylactic management of irAEs while also potentially providing a survival benefit.
Collapse
Affiliation(s)
- Michael Dougan
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA; (M.D.); (E.I.B.)
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA;
| | - Long H. Nguyen
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA;
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Elizabeth I. Buchbinder
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA; (M.D.); (E.I.B.)
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Hillard M. Lazarus
- Department of Medicine, Division of Hematology and Oncology, Case Western Reserve University, Cleveland, OH 44106, USA
| |
Collapse
|
9
|
Sampaio Moura N, Schledwitz A, Alizadeh M, Patil SA, Raufman JP. Matrix metalloproteinases as biomarkers and therapeutic targets in colitis-associated cancer. Front Oncol 2024; 13:1325095. [PMID: 38288108 PMCID: PMC10824561 DOI: 10.3389/fonc.2023.1325095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 12/26/2023] [Indexed: 01/31/2024] Open
Abstract
Colorectal cancer (CRC) remains a major cause of morbidity and mortality. Therapeutic approaches for advanced CRC are limited and rarely provide long-term benefit. Enzymes comprising the 24-member matrix metalloproteinase (MMP) family of zinc- and calcium-dependent endopeptidases are key players in extracellular matrix degradation, a requirement for colon tumor expansion, invasion, and metastasis; hence, MMPs are an important research focus. Compared to sporadic CRC, less is known regarding the molecular mechanisms and the role of MMPs in the development and progression of colitis-associated cancer (CAC) - CRC on a background of chronic inflammatory bowel disease (IBD) - primarily ulcerative colitis and Crohn's disease. Hence, the potential of MMPs as biomarkers and therapeutic targets for CAC is uncertain. Our goal was to review data regarding the role of MMPs in the development and progression of CAC. We sought to identify promising prognostic and therapeutic opportunities and novel lines of investigation. A key observation is that since MMPs may be more active in early phases of CAC, using MMPs as biomarkers of advancing neoplasia and as potential therapeutic targets for adjuvant therapy in those with advanced stage primary CAC rather than overt metastases may yield more favorable outcomes.
Collapse
Affiliation(s)
- Natalia Sampaio Moura
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Alyssa Schledwitz
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Madeline Alizadeh
- The Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Seema A. Patil
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Jean-Pierre Raufman
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, MD, United States
- Medical Service, Veterans Affairs Maryland Healthcare System, Baltimore, MD, United States
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland Medical Center, Baltimore, MD, United States
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
10
|
Hu M, Meganathan I, Zhu J, MacArthur R, Kassiri Z. Loss of TIMP3, but not TIMP4, exacerbates thoracic and abdominal aortic aneurysm. J Mol Cell Cardiol 2023; 184:61-74. [PMID: 37844423 DOI: 10.1016/j.yjmcc.2023.10.001] [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: 02/13/2023] [Revised: 10/01/2023] [Accepted: 10/01/2023] [Indexed: 10/18/2023]
Abstract
AIMS Aorta exhibits regional heterogeneity (structural and functional), while different etiologies for thoracic and abdominal aortic aneurysm (TAA, AAA) are recognized. Tissue inhibitor of metalloproteinases (TIMPs) regulate vascular remodeling through different mechanisms. Region-dependent functions have been reported for TIMP3 and TIMP4 in vascular pathologies. We investigated the region-specific function of these TIMPs in development of TAA versus AAA. METHODS & RESULTS TAA or AAA was induced in male and female mice lacking TIMP3 (Timp3-/-), TIMP4 (Timp4-/-) or in wildtype (WT) mice by peri-adventitial elastase application. Loss of TIMP3 exacerbated TAA and AAA severity in males and females, with a greater increase in proteinase activity, smooth muscle cell phenotypic switching post-AAA and -TAA, while increased inflammation was detected in the media post-AAA, but in the adventitia post-TAA. Timp3-/- mice showed impaired intimal barrier integrity post-AAA, but a greater adventitial vasa-vasorum branching post-TAA, which could explain the site of inflammation in AAA versus TAA. Severity of TAA and AAA in Timp4-/- mice was similar to WT mice. In vitro, Timp3 knockdown more severely compromised the permeability of human aortic EC monolayer compared to Timp4 knockdown or the control group. In aneurysmal aorta specimens from patients, TIMP3 expression decreased in the media in AAA, and in adventitial in TAA specimens, consistent with the impact of its loss in AAA versus TAA in mice. CONCLUSION TIMP3 loss exacerbates inflammation, adverse remodeling and aortic dilation, but triggers different patterns of remodeling in AAA versus TAA, and through different mechanisms.
Collapse
Affiliation(s)
- Mei Hu
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Ilamaran Meganathan
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Jiechun Zhu
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Rodrick MacArthur
- Department of Cardiac surgery, Mazankowski Alberta Heart Institute, University of Alberta Hospital, Edmonton, AB, Canada
| | - Zamaneh Kassiri
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
11
|
Patras L, Paul D, Matei IR. Weaving the nest: extracellular matrix roles in pre-metastatic niche formation. Front Oncol 2023; 13:1163786. [PMID: 37350937 PMCID: PMC10282420 DOI: 10.3389/fonc.2023.1163786] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 05/15/2023] [Indexed: 06/24/2023] Open
Abstract
The discovery that primary tumors condition distant organ sites of future metastasis for seeding by disseminating tumor cells through a process described as the pre-metastatic niche (PMN) formation revolutionized our understanding of cancer progression and opened new avenues for therapeutic interventions. Given the inherent inefficiency of metastasis, PMN generation is crucial to ensure the survival of rare tumor cells in the otherwise hostile environments of metastatic organs. Early on, it was recognized that preparing the "soil" of the distal organ to support the outgrowth of metastatic cells is the initiating event in PMN development, achieved through the remodeling of the organ's extracellular matrix (ECM). Remote restructuring of ECM at future sites of metastasis under the influence of primary tumor-secreted factors is an iterative process orchestrated through the crosstalk between resident stromal cells, such as fibroblasts, epithelial and endothelial cells, and recruited innate immune cells. In this review, we will explore the ECM changes, cellular effectors, and the mechanisms of ECM remodeling throughout PMN progression, as well as its impact on shaping the PMN and ultimately promoting metastasis. Moreover, we highlight the clinical and translational implications of PMN ECM changes and opportunities for therapeutically targeting the ECM to hinder PMN formation.
Collapse
Affiliation(s)
- Laura Patras
- Children’s Cancer and Blood Foundation Laboratories, Department of Pediatrics, Division of Hematology/Oncology, Drukier Institute for Children’s Health, Meyer Cancer Center, Weill Cornell Medicine, New York, NY, United States
- Department of Molecular Biology and Biotechnology, Center of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, Babes-Bolyai University, Cluj-Napoca, Romania
| | - Doru Paul
- Division of Hematology/Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Irina R. Matei
- Children’s Cancer and Blood Foundation Laboratories, Department of Pediatrics, Division of Hematology/Oncology, Drukier Institute for Children’s Health, Meyer Cancer Center, Weill Cornell Medicine, New York, NY, United States
- Department of Molecular Biology and Biotechnology, Center of Systems Biology, Biodiversity and Bioresources, Faculty of Biology and Geology, Babes-Bolyai University, Cluj-Napoca, Romania
| |
Collapse
|
12
|
li M, Yan T, Cai Y, Wei Y, Xie Q. Expression of matrix metalloproteinases and their association with clinical characteristics of solid tumors. Gene X 2023; 850:146927. [DOI: 10.1016/j.gene.2022.146927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/21/2022] [Accepted: 09/26/2022] [Indexed: 11/06/2022] Open
|
13
|
Kowalczyk M, Panasiuk-Kowalczyk A, Stadnik A, Guz M, Cybulski M, Jeleniewicz W, Stepulak A, Kwiatosz-Muc M. Dexmedetomidine Increases MMP-12 and MBP Concentrations after Coronary Artery Bypass Graft Surgery with Extracorporeal Circulation Anaesthesia without Impacting Cognitive Function: A Randomised Control Trial. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:16512. [PMID: 36554397 PMCID: PMC9778911 DOI: 10.3390/ijerph192416512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/02/2022] [Accepted: 12/07/2022] [Indexed: 06/17/2023]
Abstract
Postoperative neurological deficits remain a concern for patients undergoing cardiac surgeries. Even minor injuries can lead to neurocognitive decline (i.e., postoperative cognitive dysfunction). Dexmedetomidine may be beneficial given its reported neuroprotective effect. We aimed to investigate the effects of dexmedetomidine on brain injury during cardiac surgery anaesthesia. This prospective observational study analysed data for 46 patients who underwent coronary artery bypass graft surgery with extracorporeal circulation between August 2018 and March 2019. The patients were divided into two groups: control (CON) with typical anaesthesia and dexmedetomidine (DEX) with dexmedetomidine infusion. Concentrations of the biomarkers matrix metalloproteinase-12 (MMP-12) and myelin basic protein (MBP) were measured preoperatively and at 24 and 72 h postoperatively. Cognitive evaluations were performed preoperatively, at discharge, and 3 months after discharge using Addenbrooke's Cognitive Examination version III (ACE-III). The primary endpoint was the ACE-III score at discharge. Increased MMP-12 and MBP concentrations were observed in the DEX group 24 and 72 h postoperatively. No significant differences in ACE-III scores were observed between the groups at discharge; however, the values were increased when compared with initial values after 3 months (p = 0.000). The current results indicate that the administration of dexmedetomidine as an adjuvant to anaesthesia can increase MMP-12 and MBP levels without effects on neurocognitive outcomes at discharge and 3 months postoperatively.
Collapse
Affiliation(s)
- Michał Kowalczyk
- 1st Department of Anaesthesiology and Intensive Care, Medical University of Lublin, ul. Jaczewskiego 8, 20-954 Lublin, Poland
| | - Anna Panasiuk-Kowalczyk
- 1st Department of Anaesthesiology and Intensive Care, Medical University of Lublin, ul. Jaczewskiego 8, 20-954 Lublin, Poland
| | - Adam Stadnik
- Department of Cardiac Surgery, Medical University of Lublin, ul. Jaczewskiego 8, 20-954 Lublin, Poland
| | - Małgorzata Guz
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, ul. Chodźki 1, 20-093 Lublin, Poland
| | - Marek Cybulski
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, ul. Chodźki 1, 20-093 Lublin, Poland
| | - Witold Jeleniewicz
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, ul. Chodźki 1, 20-093 Lublin, Poland
| | - Andrzej Stepulak
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, ul. Chodźki 1, 20-093 Lublin, Poland
| | - Magdalena Kwiatosz-Muc
- 1st Department of Anaesthesiology and Intensive Care, Medical University of Lublin, ul. Jaczewskiego 8, 20-954 Lublin, Poland
| |
Collapse
|
14
|
Kim EE, Youn H, Kang KW. Imaging in Tumor Immunology. Nucl Med Mol Imaging 2021; 55:225-236. [PMID: 34721715 PMCID: PMC8517056 DOI: 10.1007/s13139-021-00706-6] [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: 03/02/2021] [Revised: 05/31/2021] [Accepted: 06/15/2021] [Indexed: 10/20/2022] Open
Abstract
Recent advances in immune modulation have made impressive progress in cancer immunotherapy. Because dynamic nature of the immune response often makes it difficult to evaluate therapeutic outcomes, innovative imaging technologies have been developed to enable non-invasive visualization of immune cells and tumors in their microenvironment. This review summarizes the current tumor immunology and describes new innovative imaging methods with great potential to obtain non-invasive real-time insights into the complex functions of the immune system and into the management of cancer immunotherapy.
Collapse
Affiliation(s)
- Euishin Edmund Kim
- Department of Radiological Sciences, UCI Medical Center, Orange County, CA USA
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, South Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Seoul, South Korea
| | - Hyewon Youn
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, South Korea
- Laboratory of Molecular Imaging and Therapy, Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Keon Wook Kang
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, South Korea
- Laboratory of Molecular Imaging and Therapy, Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
15
|
The paradoxical role of matrix metalloproteinase-11 in cancer. Biomed Pharmacother 2021; 141:111899. [PMID: 34346316 DOI: 10.1016/j.biopha.2021.111899] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 06/15/2021] [Accepted: 07/01/2021] [Indexed: 02/08/2023] Open
Abstract
The microenvironment surrounding the tumor affects biological processes, such as cell proliferation, angiogenesis, apoptosis, and invasion. Therefore, the ability to change these environments is an important attribute for tumor cells to obtain specific functions necessary for growth and metastasis. Matrix metalloproteinases (MMPs) are zinc-dependent proteolytic metalloenzymes that facilitate protease-dependent tumor progression by degrading extracellular matrix (ECM) proteins, releasing cytokines, growth factors, and other cell surface molecules. As one of the most widely studied MMPs, MMP-11 is an important protease that is expressed in cancer cells, stromal cells, and the adjacent microenvironment. MMP-11 has a dual effect on tumors. On one hand, MMP-11 promotes tumor development by inhibiting apoptosis and promoting the migration and invasion of cancer cells in the early stage. On the other hand, in animal models, MMP-11 has a protective effect on tumor growth and metastasis at an advanced stage. Based on current findings regarding the importance of MMP-11 in altering the tumor microenvironment, there is a need to further understand how stromal cells and the ECM regulate tumor progression, which may result in the re-examination of MMPs as drug targets for cancer and other diseases. In this review, we summarize the dual role of MMP-11 in cancer and its potential clinical significance.
Collapse
|
16
|
Role of Matrix Metalloproteinases in Angiogenesis and Its Implications in Asthma. J Immunol Res 2021; 2021:6645072. [PMID: 33628848 PMCID: PMC7896871 DOI: 10.1155/2021/6645072] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 01/21/2021] [Accepted: 01/25/2021] [Indexed: 12/19/2022] Open
Abstract
Asthma is a chronic airway disorder associated with aberrant inflammatory and remodeling responses. Angiogenesis and associated vascular remodeling are one of the pathological hallmarks of asthma. The mechanisms underlying angiogenesis in asthmatic airways and its clinical relevance represent a relatively nascent field in asthma when compared to other airway remodeling features. Matrix metalloproteinases (MMPs) are proteases that play an important role in both physiological and pathological conditions. In addition to facilitating extracellular matrix turnover, these proteolytic enzymes cleave bioactive molecules, thereby regulating cell signaling. MMPs have been implicated in the pathogenesis of asthma by interacting with both the airway inflammatory cells and the resident structural cells. MMPs also cover a broad range of angiogenic functions, from the degradation of the vascular basement membrane and extracellular matrix remodeling to the release of a variety of angiogenic mediators and growth factors. This review focuses on the contribution of MMPs and the regulatory role exerted by them in angiogenesis and vascular remodeling in asthma as well as addresses their potential as therapeutic targets in ameliorating angiogenesis in asthma.
Collapse
|
17
|
Contreras-Salinas H, Meza-Rios A, García-Bañuelos J, Sandoval-Rodriguez A, Sanchez-Orozco L, García-Benavides L, De la Rosa-Bibiano R, Monroy Ramirez HC, Gutiérrez-Cuevas J, Santos-Garcia A, Armendariz-Borunda J. Fibrosis regression is induced by AdhMMP8 in a murine model of chronic kidney injury. PLoS One 2020; 15:e0243307. [PMID: 33275619 PMCID: PMC7717566 DOI: 10.1371/journal.pone.0243307] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 11/18/2020] [Indexed: 12/25/2022] Open
Abstract
Adenoviral vector AdhMMP8 (human Metalloproteinase-8 cDNA) administration has been proven beneficial in various experimental models of liver injury improving liver function and decreasing fibrosis. In this study, we evaluated the potential therapeutic AdhMMP8 effect in a chronic kidney damage experimental model. Chronic injury was induced by orogastric adenine administration (100mg/kg/day) to Wistar rats for 4 weeks. AdhMMP8 (3x1011vp/kg) was administrated in renal vein during an induced-ligation-ischemic period to facilitate kidney transduction causing no-additional kidney injury as determined by histology and serum creatinine. Animals were sacrificed at 7- and 14-days post-Ad injection. Fibrosis, histopathological features, serum creatinine (sCr), BUN, and renal mRNA expression of αSMA, Col-1α, TGF-β1, CTGF, BMP7, IL-1, TNFα, VEGF and PAX2 were analyzed. Interestingly, AdhMMP8 administration resulted in cognate human MMP8 protein detection in both kidneys, whereas hMMP8 mRNA was detected only in the left kidney. AdhMMP8 significantly reduced kidney tubule-interstitial fibrosis and glomerulosclerosis. Also, tubular atrophy and interstitial inflammation were clearly decreased rendering improved histopathology, and down regulation of profibrogenic genes expression. Functionally, sCr and BUN were positively modified. The results showed that AdhMMP8 decreased renal fibrosis, suggesting that MMP8 could be a possible therapeutic candidate for kidney fibrosis treatment.
Collapse
Affiliation(s)
- Homero Contreras-Salinas
- Department of Molecular Biology and Genomics, Institute for Molecular Biology in Medicine and Gene Therapy, Health Sciences University Center, University of Guadalajara, Guadalajara, Jalisco, Mexico
| | - Alejandra Meza-Rios
- School of Medicine and Health Sciences, Tecnologico de Monterrey, Monterrey, Mexico
| | - Jesús García-Bañuelos
- Department of Molecular Biology and Genomics, Institute for Molecular Biology in Medicine and Gene Therapy, Health Sciences University Center, University of Guadalajara, Guadalajara, Jalisco, Mexico
| | - Ana Sandoval-Rodriguez
- Department of Molecular Biology and Genomics, Institute for Molecular Biology in Medicine and Gene Therapy, Health Sciences University Center, University of Guadalajara, Guadalajara, Jalisco, Mexico
| | - Laura Sanchez-Orozco
- Department of Molecular Biology and Genomics, Institute for Molecular Biology in Medicine and Gene Therapy, Health Sciences University Center, University of Guadalajara, Guadalajara, Jalisco, Mexico
| | - Leonel García-Benavides
- Department of Biomedical Sciences, Tonala University Center, University of Guadalajara, Tonala, Jalisco, Mexico
| | - Ricardo De la Rosa-Bibiano
- Department of Molecular Biology and Genomics, Institute for Molecular Biology in Medicine and Gene Therapy, Health Sciences University Center, University of Guadalajara, Guadalajara, Jalisco, Mexico
| | - Hugo Christian Monroy Ramirez
- Department of Molecular Biology and Genomics, Institute for Molecular Biology in Medicine and Gene Therapy, Health Sciences University Center, University of Guadalajara, Guadalajara, Jalisco, Mexico
| | - Jorge Gutiérrez-Cuevas
- Department of Molecular Biology and Genomics, Institute for Molecular Biology in Medicine and Gene Therapy, Health Sciences University Center, University of Guadalajara, Guadalajara, Jalisco, Mexico
| | - Arturo Santos-Garcia
- School of Medicine and Health Sciences, Tecnologico de Monterrey, Monterrey, Mexico
| | - Juan Armendariz-Borunda
- Department of Molecular Biology and Genomics, Institute for Molecular Biology in Medicine and Gene Therapy, Health Sciences University Center, University of Guadalajara, Guadalajara, Jalisco, Mexico
- School of Medicine and Health Sciences, Tecnologico de Monterrey, Monterrey, Mexico
| |
Collapse
|
18
|
Soleimany AP, Kirkpatrick JD, Su S, Dudani JS, Zhong Q, Bekdemir A, Bhatia SN. Activatable Zymography Probes Enable In Situ Localization of Protease Dysregulation in Cancer. Cancer Res 2020; 81:213-224. [PMID: 33106334 DOI: 10.1158/0008-5472.can-20-2410] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/16/2020] [Accepted: 10/21/2020] [Indexed: 12/25/2022]
Abstract
Recent years have seen the emergence of conditionally activated diagnostics and therapeutics that leverage protease-cleavable peptide linkers to enhance their specificity for cancer. However, due to a lack of methods to measure and localize protease activity directly within the tissue microenvironment, the design of protease-activated agents has been necessarily empirical, yielding suboptimal results when translated to patients. To address the need for spatially resolved protease activity profiling in cancer, we developed a new class of in situ probes that can be applied to fresh-frozen tissue sections in a manner analogous to immunofluorescence staining. These activatable zymography probes (AZP) detected dysregulated protease activity in human prostate cancer biopsy samples, enabling disease classification. AZPs were leveraged within a generalizable framework to design conditional cancer diagnostics and therapeutics and showcased in the Hi-Myc mouse model of prostate cancer, which models features of early pathogenesis. Multiplexed screening against barcoded substrates yielded a peptide, S16, that was robustly and specifically cleaved by tumor-associated metalloproteinases in the Hi-Myc model. In situ labeling with an AZP incorporating S16 revealed a potential role of metalloproteinase dysregulation in proliferative, premalignant Hi-Myc prostatic glands. Systemic administration of an in vivo imaging probe incorporating S16 perfectly classified diseased and healthy prostates, supporting the relevance of ex vivo activity assays to in vivo translation. We envision AZPs will enable new insights into the biology of protease dysregulation in cancer and accelerate the development of conditional diagnostics and therapeutics for multiple cancer types. SIGNIFICANCE: Visualization of protease activity within the native tissue context using AZPs provides new biological insights into protease dysregulation in cancer and guides the design of conditional diagnostics and therapeutics.
Collapse
Affiliation(s)
- Ava P Soleimany
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Harvard-MIT Division of Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Harvard Graduate Program in Biophysics, Harvard University, Boston, Massachusetts
| | - Jesse D Kirkpatrick
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Harvard-MIT Division of Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Susan Su
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Jaideep S Dudani
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Qian Zhong
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Ahmet Bekdemir
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Harvard-MIT Division of Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Sangeeta N Bhatia
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts. .,Harvard-MIT Division of Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Howard Hughes Medical Institute, Cambridge, Massachusetts.,Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.,Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts.,Wyss Institute at Harvard, Boston, Massachusetts.,Ludwig Center at the MIT Koch Institute for Integrative Cancer Research, Cambridge, Massachusetts
| |
Collapse
|
19
|
Fu H, Sun Y, Shao Y, Saredy J, Cueto R, Liu L, Drummer C, Johnson C, Xu K, Lu Y, Li X, Meng S, Xue ER, Tan J, Jhala NC, Yu D, Zhou Y, Bayless KJ, Yu J, Rogers TJ, Hu W, Snyder NW, Sun J, Qin X, Jiang X, Wang H, Yang X. Interleukin 35 Delays Hindlimb Ischemia-Induced Angiogenesis Through Regulating ROS-Extracellular Matrix but Spares Later Regenerative Angiogenesis. Front Immunol 2020; 11:595813. [PMID: 33154757 PMCID: PMC7591706 DOI: 10.3389/fimmu.2020.595813] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 09/22/2020] [Indexed: 12/12/2022] Open
Abstract
Interleukin (IL) 35 is a novel immunosuppressive heterodimeric cytokine in IL-12 family. Whether and how IL-35 regulates ischemia-induced angiogenesis in peripheral artery diseases are unrevealed. To fill this important knowledge gap, we used loss-of-function, gain-of-function, omics data analysis, RNA-Seq, in vivo and in vitro experiments, and we have made the following significant findings: i) IL-35 and its receptor subunit IL-12RB2, but not IL-6ST, are induced in the muscle after hindlimb ischemia (HLI); ii) HLI-induced angiogenesis is improved in Il12rb2-/- mice, in ApoE-/-/Il12rb2-/- mice compared to WT and ApoE-/- controls, respectively, where hyperlipidemia inhibits angiogenesis in vivo and in vitro; iii) IL-35 cytokine injection as a gain-of-function approach delays blood perfusion recovery at day 14 after HLI; iv) IL-35 spares regenerative angiogenesis at the late phase of HLI recovery after day 14 of HLI; v) Transcriptome analysis of endothelial cells (ECs) at 14 days post-HLI reveals a disturbed extracellular matrix re-organization in IL-35-injected mice; vi) IL-35 downregulates three reactive oxygen species (ROS) promoters and upregulates one ROS attenuator, which may functionally mediate IL-35 upregulation of anti-angiogenic extracellular matrix proteins in ECs; and vii) IL-35 inhibits human microvascular EC migration and tube formation in vitro mainly through upregulating anti-angiogenic extracellular matrix-remodeling proteins. These findings provide a novel insight on the future therapeutic potential of IL-35 in suppressing ischemia/inflammation-triggered inflammatory angiogenesis at early phase but sparing regenerative angiogenesis at late phase.
Collapse
Affiliation(s)
- Hangfei Fu
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Yu Sun
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Ying Shao
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Jason Saredy
- Centers for Metabolic Disease Research, Cardiovascular Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Ramon Cueto
- Centers for Metabolic Disease Research, Cardiovascular Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Lu Liu
- Centers for Metabolic Disease Research, Cardiovascular Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Charles Drummer
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Candice Johnson
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Keman Xu
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Yifan Lu
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Xinyuan Li
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Shu Meng
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | - Eric R Xue
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Judy Tan
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Nirag C Jhala
- Department of Pathology & Laboratory Medicine Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Daohai Yu
- Department of Clinical Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Yan Zhou
- Biostatistics and Bioinformatics Facility, Fox Chase Cancer Center, Temple Health, Philadelphia, PA, United States
| | - Kayla J Bayless
- Department of Molecular and Cellular Medicine, Texas A&M University College of Medicine, College Station, TX, United States
| | - Jun Yu
- Centers for Metabolic Disease Research, Cardiovascular Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Thomas J Rogers
- Center for Inflammation, Translational and Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Wenhui Hu
- Centers for Metabolic Disease Research, Cardiovascular Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Nathaniel W Snyder
- Centers for Metabolic Disease Research, Cardiovascular Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Jianxin Sun
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia, PA, United States
| | - Xuebin Qin
- National Primate Research Center, Tulane University, Covington, LA, United States
| | - Xiaohua Jiang
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States.,Centers for Metabolic Disease Research, Cardiovascular Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States.,Center for Inflammation, Translational and Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Hong Wang
- Centers for Metabolic Disease Research, Cardiovascular Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Xiaofeng Yang
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States.,Centers for Metabolic Disease Research, Cardiovascular Research, Thrombosis Research, Departments of Pharmacology, Microbiology and Immunology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States.,Center for Inflammation, Translational and Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| |
Collapse
|
20
|
Tykhomyrov AA, Zhernosekov DD, Grinenko TV. Plasminogen modulates formation and release of platelet angiogenic regulators. UKRAINIAN BIOCHEMICAL JOURNAL 2020. [DOI: 10.15407/ubj92.01.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
21
|
Javadian M, Gharibi T, Shekari N, Abdollahpour‐Alitappeh M, Mohammadi A, Hossieni A, Mohammadi H, Kazemi T. The role of microRNAs regulating the expression of matrix metalloproteinases (MMPs) in breast cancer development, progression, and metastasis. J Cell Physiol 2018; 234:5399-5412. [DOI: 10.1002/jcp.27445] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 08/28/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Mahsa Javadian
- Immunology Research Center, Tabriz University of Medical Science Tabriz Iran
- Department of Immunology Faculty of Medicine, Tabriz University of Medical Science Tabriz Iran
- Student Research Committee, Tabriz University of Medical Sciences Tabriz Iran
| | - Tohid Gharibi
- Immunology Research Center, Tabriz University of Medical Science Tabriz Iran
- Department of Immunology Faculty of Medicine, Tabriz University of Medical Science Tabriz Iran
| | - Najibeh Shekari
- Immunology Research Center, Tabriz University of Medical Science Tabriz Iran
- Department of Immunology Faculty of Medicine, Tabriz University of Medical Science Tabriz Iran
| | | | - Ali Mohammadi
- Immunology Research Center, Tabriz University of Medical Science Tabriz Iran
- Department of Immunology Faculty of Medicine, Tabriz University of Medical Science Tabriz Iran
| | - Arezoo Hossieni
- Immunology Research Center, Tabriz University of Medical Science Tabriz Iran
- Department of Immunology Faculty of Medicine, Tabriz University of Medical Science Tabriz Iran
| | - Hamed Mohammadi
- Immunology Research Center, Tabriz University of Medical Science Tabriz Iran
- Department of Immunology Faculty of Medicine, Tabriz University of Medical Science Tabriz Iran
| | - Tohid Kazemi
- Immunology Research Center, Tabriz University of Medical Science Tabriz Iran
- Department of Immunology Faculty of Medicine, Tabriz University of Medical Science Tabriz Iran
| |
Collapse
|
22
|
Allen H, Shraga-Heled N, Blumenfeld M, Dego-Ashto T, Fuchs-Telem D, Gilert A, Aberman Z, Ofir R. Human Placental-Derived Adherent Stromal Cells Co-Induced with TNF-α and IFN-γ Inhibit Triple-Negative Breast Cancer in Nude Mouse Xenograft Models. Sci Rep 2018; 8:670. [PMID: 29330447 PMCID: PMC5766494 DOI: 10.1038/s41598-017-18428-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 12/11/2017] [Indexed: 12/26/2022] Open
Abstract
Culturing 3D-expanded human placental-derived adherent stromal cells (ASCs) in the presence of tumor necrosis factor-alpha (TNF-α) and interferon-gamma (IFN-γ) transiently upregulated the secretion of numerous anti-proliferative, anti-angiogenic and pro-inflammatory cytokines. In a 3D-spheroid screening assay, conditioned medium from these induced-ASCs inhibited proliferation of cancer cell lines, including triple-negative breast cancer (TNBC) lines. In vitro co-culture studies of induced-ASCs with MDA-MB-231 human breast carcinoma cells, a model representing TNBC, supports a mechanism involving immunomodulation and angiogenesis inhibition. In vivo studies in nude mice showed that intramuscular administration of induced-ASCs halted MDA-MB-231 cell proliferation, and inhibited tumor progression and vascularization. Thirty percent of treated mice experienced complete tumor remission. Murine serum concentrations of the tumor-supporting cytokines Interleukin-6 (IL-6), Vascular endothelial growth factor (VEGF) and Granulocyte-colony stimulating factor (G-CSF) were lowered to naïve levels. A somatic mutation analysis identified numerous genes which could be screened in patients to increase a positive therapeutic outcome. Taken together, these results show that targeted changes in the secretion profile of ASCs may improve their therapeutic potential.
Collapse
|
23
|
Han F, Zhang S, Zhang L, Hao Q. The overexpression and predictive significance of MMP-12 in esophageal squamous cell carcinoma. Pathol Res Pract 2017; 213:1519-1522. [PMID: 29033183 DOI: 10.1016/j.prp.2017.09.023] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 09/18/2017] [Accepted: 09/20/2017] [Indexed: 11/17/2022]
Abstract
Matrix metalloproteinase (MMP)-mediated degradation of the extracellular matrix is a major factor for tumor invasion and metastasis. MMP-12, as metalloelastase, its function in tumor progression remains contradictory. This study was undertaken to investigate the role of MMP-12 in esophageal squamous cell carcinoma (ESCC). We analyzed the protein expression of MMP-12 and its association with clinicopatholigical parameters, as well as survival analysis. MMP-12 was highly expressed in tumor cells comparing with normal epithelial cells. The high expression of MMP-12 was significantly correlated with tumor grade and stage, nodal metastasis and poor survival of ESCC. Cox multivariate analysis revealed that MMP-12 was an independent prognostic factor in ESCC. Our results suggest that MMP-12 might act as a potential target for the development of novel therapeutics of esophageal squamous cell carcinoma.
Collapse
Affiliation(s)
- Fei Han
- Division of Radiological Sciences, Washington University School of Medicine in Saint Louis, MO, 63110, USA; Department of Pathology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China.
| | - Suxia Zhang
- Department of Pathology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Long Zhang
- Department of Pathology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Qiongyu Hao
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| |
Collapse
|
24
|
Merchant N, Nagaraju GP, Rajitha B, Lammata S, Jella KK, Buchwald ZS, Lakka SS, Ali AN. Matrix metalloproteinases: their functional role in lung cancer. Carcinogenesis 2017. [DOI: 10.1093/carcin/bgx063] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
25
|
Spitler LE, Cao H, Piironen T, Whiteside TL, Weber RW, Cruickshank S. Biological Effects of Anti-Granulocyte-Macrophage Colony-Stimulating Factor (GM-CSF) Antibody Formation in Patients Treated With GM-CSF (Sargramostim) as Adjuvant Therapy of Melanoma. Am J Clin Oncol 2017; 40:207-213. [PMID: 25286079 PMCID: PMC4385005 DOI: 10.1097/coc.0000000000000124] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES We investigated the development of binding and neutralizing antibodies to granulocyte-macrophage colony-stimulating factor (GM-CSF) in patients receiving prolonged therapy with GM-CSF as adjuvant therapy of melanoma and the impact of these antibodies on biological effects. METHODS Fifty-three patients with high-risk melanoma that had been surgically excised were treated with GM-CSF, 125 μg/m daily for 14 days every 28 days for 1 year after surgical resection of disease. Serum samples for antibodies to GM-CSF were measured before treatment and on study days 155 and 351. Blood draws for testing biological effects were keyed to GM-CSF administration: days 0 (before), 15 (after 14 d on GM-CSF), 29 (after 14 d off GM-CSF), 155, and 351 (after 14 d on GM-CSF in the sixth and 13th cycle of treatment). RESULTS Of 53 patients enrolled, 43 were evaluable for the development of anti-GM-CSF antibodies. Of these, 93% developed binding antibodies and 42% developed both binding and neutralizing antibodies. The increase in the white blood cell count, percent eosinophils, or neopterin levels engendered by GM-CSF administration was abrogated or markedly decreased in patients with neutralizing antibodies but not in patients who developed only binding antibodies. CONCLUSIONS Ninety-three percent of patients with melanoma treated with GM-CSF as adjuvant therapy develop antibodies to GM-CSF. In those with neutralizing antibodies, a diminution of the biological effects of GM-CSF was observed. The development of neutralizing antibodies might also abrogate the potential clinical benefit of this treatment and should be considered in the design of future clinical trials.
Collapse
Affiliation(s)
- Lynn E. Spitler
- Northern California Melanoma Center, St. Mary’s Medical Center, San Francisco, CA
| | - Huynh Cao
- Northern California Melanoma Center, St. Mary’s Medical Center, San Francisco, CA
| | | | | | - Robert W. Weber
- Northern California Melanoma Center, St. Mary’s Medical Center, San Francisco, CA
| | | |
Collapse
|
26
|
Chelluboina B, Nalamolu KR, Klopfenstein JD, Pinson DM, Wang DZ, Vemuganti R, Veeravalli KK. MMP-12, a Promising Therapeutic Target for Neurological Diseases. Mol Neurobiol 2017; 55:1405-1409. [PMID: 28155200 DOI: 10.1007/s12035-017-0418-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 01/24/2017] [Indexed: 12/01/2022]
Abstract
The role of matrix metalloproteinase-12 (MMP-12) in the pathogenesis of several inflammatory diseases such as chronic obstructive pulmonary disease, emphysema, and asthma is well established. Several new studies and recent reports from our laboratory and others highlighted the detrimental role of MMP-12 in the pathogenesis of several neurological diseases. In this review, we discuss in detail the pathological role of MMP-12 and the possible underlying molecular mechanisms that contribute to disease pathogenesis in the context of central nervous system diseases such as stroke, spinal cord injury, and multiple sclerosis. The available information on the specific MMP-12 inhibitors used in several preclinical and clinical studies is also reviewed. Based on the reported studies to date, MMP-12 suppression could emerge as a promising therapeutic target for several CNS diseases that were discussed in this review.
Collapse
Affiliation(s)
- Bharath Chelluboina
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, One Illini Dr., Peoria, IL, 61605, USA
| | - Koteswara Rao Nalamolu
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, One Illini Dr., Peoria, IL, 61605, USA
| | - Jeffrey D Klopfenstein
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, One Illini Dr., Peoria, IL, 61605, USA.,Department of Neurosurgery, University of Illinois College of Medicine at Peoria, Peoria, IL, USA.,Comprehensive Stroke Center, Illinois Neurological Institute, Peoria, IL, USA
| | - David M Pinson
- Department of Pathology, University of Illinois College of Medicine at Peoria, Peoria, IL, USA
| | - David Z Wang
- Comprehensive Stroke Center, Illinois Neurological Institute, Peoria, IL, USA.,Department of Neurology, University of Illinois College of Medicine at Peoria, Peoria, IL, USA
| | - Raghu Vemuganti
- Department of Neurological Surgery, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Krishna Kumar Veeravalli
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, One Illini Dr., Peoria, IL, 61605, USA. .,Department of Neurosurgery, University of Illinois College of Medicine at Peoria, Peoria, IL, USA. .,Department of Neurology, University of Illinois College of Medicine at Peoria, Peoria, IL, USA.
| |
Collapse
|
27
|
Acceleration of tumor growth due to dysfunction in M1 macrophages and enhanced angiogenesis in an animal model of autoimmune disease. J Transl Med 2016; 96:468-80. [PMID: 26808709 DOI: 10.1038/labinvest.2015.166] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 12/04/2015] [Accepted: 12/16/2015] [Indexed: 01/15/2023] Open
Abstract
Both autoimmunity and tumor immunity are immune responses against self-tissues or cells. However, the precise similarity or difference between them remains unclear. In this study, to understand a novel mechanism of tumor immunity, we performed transplantation experiments with a murine autoimmune model, C57BL/6J (B6)/lpr mice. A melanoma cell line, B16F10 cells, or granulocyte macrophage colony-stimulating factor- overexpressing B16F10 (B16F10/mGM) cells were transplanted into B6 or B6/lpr mice. Tumor growth by transplanted B16F10/mGM cells was significantly accelerated in B6/lpr mice compared with that in B6 mice. The accumulation of M1 macrophages in the tumor tissues of B6/lpr recipient mice was significantly lower compared with that in the control mice. In vitro co-culture experiment showed that impaired differentiation into M1 macrophages was observed in B6/lpr mice. The number of tumor vessels and vascular endothelial growth factor (VEGF) expression were also significantly enhanced in the tumor tissues of B6/lpr mice compared with those in the B6 mice. Moreover, VEGF expression was correlated with the increased expression of hypoxia-inducible factor-1α in the tumor tissues of B6/lpr mice. These results suggest that dysfunctional tumor immunity and enhanced angiogenesis in autoimmunity influence tumor growth.
Collapse
|
28
|
Schauer D, Starlinger P, Alidzanovic L, Zajc P, Maier T, Feldman A, Padickakudy R, Buchberger E, Elleder V, Spittler A, Stift J, Pop L, Gruenberger B, Gruenberger T, Brostjan C. Chemotherapy of colorectal liver metastases induces a rapid rise in intermediate blood monocytes which predicts treatment response. Oncoimmunology 2016; 5:e1160185. [PMID: 27471631 DOI: 10.1080/2162402x.2016.1160185] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 02/15/2016] [Accepted: 02/25/2016] [Indexed: 12/13/2022] Open
Abstract
We have previously reported that intermediate monocytes (CD14(++)/CD16(+)) were increased in colorectal cancer (CRC) patients, while the subset of pro-angiogenic TIE2-expressing monocytes (TEMs) was not significantly elevated. This study was designed to evaluate changes in frequency and function of intermediate monocytes and TEMs during chemotherapy and anti-angiogenic cancer treatment and their relation to treatment response. Monocyte populations were determined by flow cytometry in 60 metastasized CRC (mCRC) patients who received neoadjuvant chemotherapy with or without bevacizumab. Blood samples were taken before treatment, after two therapy cycles, at the end of neoadjuvant therapy and immediately before surgical resection of liver metastases. Neoadjuvant treatment resulted in a significant increase in circulating intermediate monocytes which was most pronounced after two cycles and positively predicted tumor response (AUC = 0.875, p = 0.005). With a cut-off value set to 1% intermediate monocytes of leukocytes, this parameter showed a predictive sensitivity and specificity of 75% and 88%. Anti-angiogenic therapy with bevacizumab had no impact on monocyte populations including TEMs. In 15 patients and six healthy controls, the gene expression profile and the migratory behavior of monocyte subsets was evaluated. The profile of intermediate monocytes suggested functions in antigen presentation, inflammatory cytokine production, chemotaxis and was remarkably stable during chemotherapy. Intermediate monocytes showed a preferential migratory response to tumor-derived signals in vitro and correlated with the level of CD14(+)/CD16(+) monocytic infiltrates in the resected tumor tissue. In conclusion, the rapid rise of intermediate monocytes during chemotherapy may offer a simple marker for response prediction and a timely change in regimen.
Collapse
Affiliation(s)
- Dominic Schauer
- Department of Surgery, Medical University of Vienna, General Hospital, Waehringer Guertel , Vienna, Austria
| | - Patrick Starlinger
- Department of Surgery, Medical University of Vienna, General Hospital, Waehringer Guertel , Vienna, Austria
| | - Lejla Alidzanovic
- Department of Surgery, Medical University of Vienna, General Hospital, Waehringer Guertel , Vienna, Austria
| | - Philipp Zajc
- Department of Surgery, Medical University of Vienna, General Hospital, Waehringer Guertel , Vienna, Austria
| | - Thomas Maier
- Department of Surgery, Medical University of Vienna, General Hospital, Waehringer Guertel , Vienna, Austria
| | - Alexandra Feldman
- Department of Surgery, Medical University of Vienna, General Hospital, Waehringer Guertel , Vienna, Austria
| | - Robin Padickakudy
- Department of Surgery, Medical University of Vienna, General Hospital, Waehringer Guertel , Vienna, Austria
| | - Elisabeth Buchberger
- Department of Surgery, Medical University of Vienna, General Hospital, Waehringer Guertel , Vienna, Austria
| | - Vanessa Elleder
- Department of Surgery, Medical University of Vienna, General Hospital, Waehringer Guertel , Vienna, Austria
| | - Andreas Spittler
- Department of Surgery, Medical University of Vienna, General Hospital, Waehringer Guertel, Vienna, Austria; Core Facility Flow Cytometry, Medical University of Vienna, General Hospital, Waehringer Guertel, Vienna, Austria
| | - Judith Stift
- Department of Pathology, Medical University of Vienna, General Hospital, Waehringer Guertel , Vienna, Austria
| | - Lorand Pop
- Department of Surgery, Medical University of Vienna, General Hospital, Waehringer Guertel , Vienna, Austria
| | - Birgit Gruenberger
- Department of Internal Medicine, Hospital of the Merciful Brothers , Johannes von Gott Platz 1 , Vienna, Austria
| | - Thomas Gruenberger
- Department of Surgery, Medical University of Vienna, General Hospital, Waehringer Guertel, Vienna, Austria; Department of Surgery I, Rudolf Foundation Clinic, Juchgasse, Vienna, Austria
| | - Christine Brostjan
- Department of Surgery, Medical University of Vienna, General Hospital, Waehringer Guertel, Vienna, Austria; Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel, Vienna, Austria
| |
Collapse
|
29
|
Bonnefont-Rebeix C, Fournel-Fleury C, Ponce F, Belluco S, Watrelot D, Bouteille SE, Rapiteau S, Razanajaona-Doll D, Pin JJ, Leroux C, Marchal T. Characterization of a novel canine T-cell line established from a spontaneously occurring aggressive T-cell lymphoma with large granular cell morphology. Immunobiology 2016; 221:12-22. [DOI: 10.1016/j.imbio.2015.08.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 06/15/2015] [Accepted: 08/11/2015] [Indexed: 11/17/2022]
|
30
|
Parris GE. A Hypothesis Concerning the Biphasic Dose-response of Tumors to Angiostatin and Endostatin. Dose Response 2015; 13:10.2203_dose-response.14-020.Parris. [PMID: 26675544 PMCID: PMC4674172 DOI: 10.2203/dose-response.14-020.parris] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
This manuscript proposes a hypothesis to explain the U-shaped dose-response observed for angiostatin and other high-molecular-weight drugs in various anti-cancer bio-assays. The dose-response curves for angiostatin and endostatin (measured as suppression of tumor growth) go through an optimum (i.e., minimum tumor growth) and then becomes less effective at higher doses. The literature suggests that at lower doses the primary action of these high-molecular-weight drugs is to counteract the angiogenic effects of vascular endothelial growth factor (VEGF). To do this, the drugs must pass out of the blood vessel and enter the extra-cellular matrix (ECM) where VEGF induces the growth and fusion of tip cells. Ironically, VEGF actually facilitates access of the drugs to the ECM by making the vascular endothelium leaky. At higher doses, the high-molecular-weight drugs seem to reverse VEGF-induced permeability of the endothelium. Thus, at high dose rates, it is hypothesized that the drugs are not able to enter the ECM and block the angiogenic effects of VEGF there. As a result, high doses of the drugs do not suppress vascularization of the tumor or tumor growth. Moreover, if the permeability of the vessels is suppressed, the VEGF released by the stroma is concentrated in the ECM where it amplifies the angiogenic activity around the tumor.
Collapse
Affiliation(s)
- George E Parris
- Montgomery College, Department of Chemistry, Rockville, MD, USA
| |
Collapse
|
31
|
Lawson DH, Lee S, Zhao F, Tarhini AA, Margolin KA, Ernstoff MS, Atkins MB, Cohen GI, Whiteside TL, Butterfield LH, Kirkwood JM. Randomized, Placebo-Controlled, Phase III Trial of Yeast-Derived Granulocyte-Macrophage Colony-Stimulating Factor (GM-CSF) Versus Peptide Vaccination Versus GM-CSF Plus Peptide Vaccination Versus Placebo in Patients With No Evidence of Disease After Complete Surgical Resection of Locally Advanced and/or Stage IV Melanoma: A Trial of the Eastern Cooperative Oncology Group-American College of Radiology Imaging Network Cancer Research Group (E4697). J Clin Oncol 2015; 33:4066-76. [PMID: 26351350 PMCID: PMC4669592 DOI: 10.1200/jco.2015.62.0500] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE We conducted a double-blind, placebo-controlled trial to evaluate the effect of granulocyte-macrophage colony-stimulating factor (GM-CSF) and peptide vaccination (PV) on relapse-free survival (RFS) and overall survival (OS) in patients with resected high-risk melanoma. PATIENTS AND METHODS Patients with completely resected stage IV or high-risk stage III melanoma were grouped by human leukocyte antigen (HLA) -A2 status. HLA-A2-positive patients were randomly assigned to receive GM-CSF, PV, both, or placebo; HLA-A2-negative patients, GM-CSF or placebo. Treatment lasted for 1 year or until recurrence. Efficacy analyses were conducted in the intent-to-treat population. RESULTS A total of 815 patients were enrolled. There were no significant improvements in OS (stratified log-rank P = .528; hazard ratio, 0.94; 95% repeated CI, 0.77 to 1.15) or RFS (P = .131; hazard ratio, 0.88; 95% CI, 0.74 to 1.04) in the patients assigned to GM-CSF (n = 408) versus those assigned to placebo (n = 407). The median OS times with GM-CSF versus placebo treatments were 69.6 months (95% CI, 53.4 to 83.5 months) versus 59.3 months (95% CI, 44.4 to 77.3 months); the 5-year OS probability rates were 52.3% (95% CI, 47.3% to 57.1%) versus 49.4% (95% CI, 44.3% to 54.3%), respectively. The median RFS times with GM-CSF versus placebo were 11.4 months (95% CI, 9.4 to 14.8 months) versus 8.8 months (95% CI, 7.5 to 11.2 months); the 5-year RFS probability rates were 31.2% (95% CI, 26.7% to 35.9%) versus 27.0% (95% CI, 22.7% to 31.5%), respectively. Exploratory analyses showed a trend toward improved OS in GM-CSF-treated patients with resected visceral metastases. When survival in HLA-A2-positive patients who received PV versus placebo was compared, RFS and OS were not significantly different. Treatment-related grade 3 or greater adverse events were similar between GM-CSF and placebo groups. CONCLUSION Neither adjuvant GM-CSF nor PV significantly improved RFS or OS in patients with high-risk resected melanoma. Exploratory analyses suggest that GM-CSF may be beneficial in patients with resected visceral metastases; this observation requires prospective validation.
Collapse
Affiliation(s)
- David H Lawson
- David H. Lawson, Winship Cancer Institute of Emory University, Atlanta, GA; Sandra Lee and Fengmin Zhao, Dana-Farber Cancer Institute; Michael B. Atkins, Beth Israel Deaconess Medical Center, Boston, MA; Ahmad A. Tarhini, Theresa L. Whiteside, Lisa H. Butterfield, and John M. Kirkwood, University of Pittsburgh Medical Center, Pittsburgh, PA; Kim A. Margolin, Seattle Cancer Care Alliance, Seattle, WA; Marc S. Ernstoff, Dartmouth-Hitchcock Medical Center, Lebanon, NH; and Gary I. Cohen, Greater Baltimore Medical Center, Baltimore, MD.
| | - Sandra Lee
- David H. Lawson, Winship Cancer Institute of Emory University, Atlanta, GA; Sandra Lee and Fengmin Zhao, Dana-Farber Cancer Institute; Michael B. Atkins, Beth Israel Deaconess Medical Center, Boston, MA; Ahmad A. Tarhini, Theresa L. Whiteside, Lisa H. Butterfield, and John M. Kirkwood, University of Pittsburgh Medical Center, Pittsburgh, PA; Kim A. Margolin, Seattle Cancer Care Alliance, Seattle, WA; Marc S. Ernstoff, Dartmouth-Hitchcock Medical Center, Lebanon, NH; and Gary I. Cohen, Greater Baltimore Medical Center, Baltimore, MD
| | - Fengmin Zhao
- David H. Lawson, Winship Cancer Institute of Emory University, Atlanta, GA; Sandra Lee and Fengmin Zhao, Dana-Farber Cancer Institute; Michael B. Atkins, Beth Israel Deaconess Medical Center, Boston, MA; Ahmad A. Tarhini, Theresa L. Whiteside, Lisa H. Butterfield, and John M. Kirkwood, University of Pittsburgh Medical Center, Pittsburgh, PA; Kim A. Margolin, Seattle Cancer Care Alliance, Seattle, WA; Marc S. Ernstoff, Dartmouth-Hitchcock Medical Center, Lebanon, NH; and Gary I. Cohen, Greater Baltimore Medical Center, Baltimore, MD
| | - Ahmad A Tarhini
- David H. Lawson, Winship Cancer Institute of Emory University, Atlanta, GA; Sandra Lee and Fengmin Zhao, Dana-Farber Cancer Institute; Michael B. Atkins, Beth Israel Deaconess Medical Center, Boston, MA; Ahmad A. Tarhini, Theresa L. Whiteside, Lisa H. Butterfield, and John M. Kirkwood, University of Pittsburgh Medical Center, Pittsburgh, PA; Kim A. Margolin, Seattle Cancer Care Alliance, Seattle, WA; Marc S. Ernstoff, Dartmouth-Hitchcock Medical Center, Lebanon, NH; and Gary I. Cohen, Greater Baltimore Medical Center, Baltimore, MD
| | - Kim A Margolin
- David H. Lawson, Winship Cancer Institute of Emory University, Atlanta, GA; Sandra Lee and Fengmin Zhao, Dana-Farber Cancer Institute; Michael B. Atkins, Beth Israel Deaconess Medical Center, Boston, MA; Ahmad A. Tarhini, Theresa L. Whiteside, Lisa H. Butterfield, and John M. Kirkwood, University of Pittsburgh Medical Center, Pittsburgh, PA; Kim A. Margolin, Seattle Cancer Care Alliance, Seattle, WA; Marc S. Ernstoff, Dartmouth-Hitchcock Medical Center, Lebanon, NH; and Gary I. Cohen, Greater Baltimore Medical Center, Baltimore, MD
| | - Marc S Ernstoff
- David H. Lawson, Winship Cancer Institute of Emory University, Atlanta, GA; Sandra Lee and Fengmin Zhao, Dana-Farber Cancer Institute; Michael B. Atkins, Beth Israel Deaconess Medical Center, Boston, MA; Ahmad A. Tarhini, Theresa L. Whiteside, Lisa H. Butterfield, and John M. Kirkwood, University of Pittsburgh Medical Center, Pittsburgh, PA; Kim A. Margolin, Seattle Cancer Care Alliance, Seattle, WA; Marc S. Ernstoff, Dartmouth-Hitchcock Medical Center, Lebanon, NH; and Gary I. Cohen, Greater Baltimore Medical Center, Baltimore, MD
| | - Michael B Atkins
- David H. Lawson, Winship Cancer Institute of Emory University, Atlanta, GA; Sandra Lee and Fengmin Zhao, Dana-Farber Cancer Institute; Michael B. Atkins, Beth Israel Deaconess Medical Center, Boston, MA; Ahmad A. Tarhini, Theresa L. Whiteside, Lisa H. Butterfield, and John M. Kirkwood, University of Pittsburgh Medical Center, Pittsburgh, PA; Kim A. Margolin, Seattle Cancer Care Alliance, Seattle, WA; Marc S. Ernstoff, Dartmouth-Hitchcock Medical Center, Lebanon, NH; and Gary I. Cohen, Greater Baltimore Medical Center, Baltimore, MD
| | - Gary I Cohen
- David H. Lawson, Winship Cancer Institute of Emory University, Atlanta, GA; Sandra Lee and Fengmin Zhao, Dana-Farber Cancer Institute; Michael B. Atkins, Beth Israel Deaconess Medical Center, Boston, MA; Ahmad A. Tarhini, Theresa L. Whiteside, Lisa H. Butterfield, and John M. Kirkwood, University of Pittsburgh Medical Center, Pittsburgh, PA; Kim A. Margolin, Seattle Cancer Care Alliance, Seattle, WA; Marc S. Ernstoff, Dartmouth-Hitchcock Medical Center, Lebanon, NH; and Gary I. Cohen, Greater Baltimore Medical Center, Baltimore, MD
| | - Theresa L Whiteside
- David H. Lawson, Winship Cancer Institute of Emory University, Atlanta, GA; Sandra Lee and Fengmin Zhao, Dana-Farber Cancer Institute; Michael B. Atkins, Beth Israel Deaconess Medical Center, Boston, MA; Ahmad A. Tarhini, Theresa L. Whiteside, Lisa H. Butterfield, and John M. Kirkwood, University of Pittsburgh Medical Center, Pittsburgh, PA; Kim A. Margolin, Seattle Cancer Care Alliance, Seattle, WA; Marc S. Ernstoff, Dartmouth-Hitchcock Medical Center, Lebanon, NH; and Gary I. Cohen, Greater Baltimore Medical Center, Baltimore, MD
| | - Lisa H Butterfield
- David H. Lawson, Winship Cancer Institute of Emory University, Atlanta, GA; Sandra Lee and Fengmin Zhao, Dana-Farber Cancer Institute; Michael B. Atkins, Beth Israel Deaconess Medical Center, Boston, MA; Ahmad A. Tarhini, Theresa L. Whiteside, Lisa H. Butterfield, and John M. Kirkwood, University of Pittsburgh Medical Center, Pittsburgh, PA; Kim A. Margolin, Seattle Cancer Care Alliance, Seattle, WA; Marc S. Ernstoff, Dartmouth-Hitchcock Medical Center, Lebanon, NH; and Gary I. Cohen, Greater Baltimore Medical Center, Baltimore, MD
| | - John M Kirkwood
- David H. Lawson, Winship Cancer Institute of Emory University, Atlanta, GA; Sandra Lee and Fengmin Zhao, Dana-Farber Cancer Institute; Michael B. Atkins, Beth Israel Deaconess Medical Center, Boston, MA; Ahmad A. Tarhini, Theresa L. Whiteside, Lisa H. Butterfield, and John M. Kirkwood, University of Pittsburgh Medical Center, Pittsburgh, PA; Kim A. Margolin, Seattle Cancer Care Alliance, Seattle, WA; Marc S. Ernstoff, Dartmouth-Hitchcock Medical Center, Lebanon, NH; and Gary I. Cohen, Greater Baltimore Medical Center, Baltimore, MD
| |
Collapse
|
32
|
Kwek SS, Kahn J, Greaney SK, Lewis J, Cha E, Zhang L, Weber RW, Leonard L, Markovic SN, Fong L, Spitler LE. GM-CSF and ipilimumab therapy in metastatic melanoma: Clinical outcomes and immunologic responses. Oncoimmunology 2015; 5:e1101204. [PMID: 27141383 DOI: 10.1080/2162402x.2015.1101204] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 09/23/2015] [Accepted: 09/23/2015] [Indexed: 12/22/2022] Open
Abstract
We conducted a phase II clinical trial of anti-CTLA-4 antibody (ipilimumab) and granulocyte-macrophage colony-stimulating factor (GM-CSF) in 22 patients with metastatic melanoma and determined clinical outcomes and immunologic responses. The treatment consisted of a 3-mo induction with ipilimumab at 10 mg/kg administered every 3 weeks for four doses in combination with GM-CSF at 125 µg/m2 for 14 d beginning on the day of the ipilimumab infusion and then GM-CSF for 3 mo on the same schedule without ipilimumab. This was followed by maintenance therapy with the combination every 3 mo for up to 2 y or until disease progression or unacceptable toxicity. Blood samples for determination of immune subsets were obtained before treatment, at week 3 (end of cycle 1) and at week 6 (end of cycle 2). Blood samples were also obtained from seven subjects who were cancer-free. The immune response disease control (irDC) rate at 24 weeks was 41% and the overall response rate (ORR) was 32%. The median progression free-survival (PFS) was 3.5 mo and the median overall survival (OS) was 21.1 mo. 41% of the patients experienced Grade 3 to 4 adverse events. We conclude that this combination is safe and the results suggest the combination may be more effective than ipilimumab monotherapy. Further, the results suggest that lower levels of CD4+ effector T cells but higher levels of CD8+ T cells expressing PD-1 at pre-treatment could be a potential biomarker for disease control in patients who receive immunotherapy with ipilimumab and GM-CSF. Further trials of this combination are warranted.
Collapse
Affiliation(s)
- Serena S Kwek
- Department of Medicine, Hem/Onc Division, University of California San Francisco , San Francisco, CA, USA
| | - James Kahn
- Northern California Melanoma Center, Saint Mary's Medical Center , San Francisco, CA, USA
| | - Samantha K Greaney
- Northern California Melanoma Center, Saint Mary's Medical Center , San Francisco, CA, USA
| | - Jera Lewis
- Department of Medicine, Hem/Onc Division, University of California San Francisco , San Francisco, CA, USA
| | - Edward Cha
- Department of Medicine, Hem/Onc Division, University of California San Francisco , San Francisco, CA, USA
| | - Li Zhang
- Department of Medicine, Hem/Onc Division, University of California San Francisco , San Francisco, CA, USA
| | - Robert W Weber
- Northern California Melanoma Center, Saint Mary's Medical Center , San Francisco, CA, USA
| | - Lonnie Leonard
- Northern California Melanoma Center, Saint Mary's Medical Center , San Francisco, CA, USA
| | - Svetomir N Markovic
- Division of Hematology, Oncology Department , Mayo Clinic , Rochester, MN, USA
| | - Lawrence Fong
- Department of Medicine, Hem/Onc Division, University of California San Francisco , San Francisco, CA, USA
| | - Lynn E Spitler
- Northern California Melanoma Center, Saint Mary's Medical Center , San Francisco, CA, USA
| |
Collapse
|
33
|
Post-transcriptional inactivation of matrix metalloproteinase-12 after focal cerebral ischemia attenuates brain damage. Sci Rep 2015; 5:9504. [PMID: 25955565 PMCID: PMC5386183 DOI: 10.1038/srep09504] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2014] [Accepted: 03/10/2015] [Indexed: 12/23/2022] Open
Abstract
This study highlights the possible pathological role of MMP-12 in the context of ischemic stroke. Male rats were subjected to a two-hour middle cerebral artery occlusion (MCAO) procedure. MMP-12 shRNA expressing plasmid formulation was administered to these rats twenty-four hours after reperfusion. The results showed a predominant upregulation of MMP-12 (approximately 47, 58, 143, and 265 folds on days 1, 3, 5, 7 post-ischemia, respectively) in MCAO subjected rats. MMP-12 expression was localized to neurons, oligodendrocytes and microglia, but not astrocytes. Transcriptional inactivation of MMP-12 significantly reduced the infarct size. The percent infarct size was reduced from 62.87 ± 4.13 to 34.67 ± 5.39 after MMP-12 knockdown compared to untreated MCAO subjected rats. Expression of myelin basic protein was increased, and activity of MMP-9 was reduced in ischemic rat brains after MMP-12 knockdown. Furthermore, a significant reduction in the extent of apoptosis was noticed after MMP-12 knockdown. TNFα expression in the ipsilateral regions of MCAO-subjected rats was reduced after MMP-12 knockdown in addition to the reduced protein expression of apoptotic molecules that are downstream to TNFα signaling. Specific knockdown of MMP-12 after focal cerebral ischemia offers neuroprotection that could be mediated via reduced MMP-9 activation and myelin degradation as well as inhibition of apoptosis.
Collapse
|
34
|
Goncalves I, Bengtsson E, Colhoun HM, Shore AC, Palombo C, Natali A, Edsfeldt A, Dunér P, Fredrikson GN, Björkbacka H, Östling G, Aizawa K, Casanova F, Persson M, Gooding K, Strain D, Khan F, Looker HC, Adams F, Belch J, Pinnoli S, Venturi E, Kozakova M, Gan LM, Schnecke V, Nilsson J. Elevated Plasma Levels of MMP-12 Are Associated With Atherosclerotic Burden and Symptomatic Cardiovascular Disease in Subjects With Type 2 Diabetes. Arterioscler Thromb Vasc Biol 2015; 35:1723-31. [PMID: 25953645 DOI: 10.1161/atvbaha.115.305631] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 04/22/2015] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Matrix metalloproteinases (MMPs) degrade extracellular matrix proteins and play important roles in development and tissue repair. They have also been shown to have both protective and pathogenic effects in atherosclerosis, and experimental studies have suggested that MMP-12 contributes to plaque growth and destabilization. The objective of this study was to investigate the associations between circulating MMPs, atherosclerosis burden, and incidence of cardiovascular disease with a particular focus on type 2 diabetes mellitus. APPROACH AND RESULTS Plasma levels of MMP-1, -3, -7, -10, and -12 were analyzed by the Proximity Extension Assay technology in 1500 subjects participating in the SUMMIT (surrogate markers for micro- and macrovascular hard end points for innovative diabetes tools) study, 384 incident coronary cases, and 409 matched controls in the Malmö Diet and Cancer study and in 205 carotid endarterectomy patients. Plasma MMP-7 and -12 were higher in subjects with type 2 diabetes mellitus, increased with age and impaired renal function, and was independently associated with prevalent cardiovascular disease, atherosclerotic burden (as assessed by carotid intima-media thickness and ankle-brachial pressure index), arterial stiffness, and plaque inflammation. Baseline MMP-7 and -12 levels were increased in Malmö Diet and Cancer subjects who had a coronary event during follow-up. CONCLUSIONS The plasma level of MMP-7 and -12 are elevated in type 2 diabetes mellitus, associated with more severe atherosclerosis and an increased incidence of coronary events. These observations provide clinical support to previous experimental studies, demonstrating a role for these MMPs in plaque development, and suggest that they are potential biomarkers of atherosclerosis burden and cardiovascular disease risk.
Collapse
Affiliation(s)
- Isabel Goncalves
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Eva Bengtsson
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Helen M Colhoun
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Angela C Shore
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Carlo Palombo
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Andrea Natali
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Andreas Edsfeldt
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Pontus Dunér
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Gunilla Nordin Fredrikson
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Harry Björkbacka
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Gerd Östling
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Kunihiko Aizawa
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Francesco Casanova
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Margaretha Persson
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Kim Gooding
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - David Strain
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Faisel Khan
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Helen C Looker
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Fiona Adams
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Jill Belch
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Silvia Pinnoli
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Elena Venturi
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Michaela Kozakova
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Li-Ming Gan
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Volker Schnecke
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.)
| | - Jan Nilsson
- From the Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden (I.G., E.B., A.E., P.D., G.N.F., H.B., G.Ö., M.P., J.N.); Medical Research Institute, University of Dundee, Dundee, UK (H.M.C., F.K., H.C.L., F.A., J.B.); Institute of Biomedical and Clinical Science, Diabetes and Vascular Medicine, NIHR Exeter Clinical Research Facility and University of Exeter Medical School, Exeter, UK (A.C.S., K.A., F.C., K.G., D.S.); Department of Clinical and Experimental Medicine (A.N., S.P., E.V.) and Department of Surgical, Medical, Molecular and Critical Area Pathology (C.P., M.K.), University of Pisa, Pisa, Italy; and AstraZeneca, Cardiovascular and Metabolic Diseases, Mölndal, Sweden (L.-M.G., V.S.).
| | | |
Collapse
|
35
|
Multifunctional bioscaffolds for 3D culture of melanoma cells reveal increased MMP activity and migration with BRAF kinase inhibition. Proc Natl Acad Sci U S A 2015; 112:5366-71. [PMID: 25870264 DOI: 10.1073/pnas.1505662112] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Matrix metalloproteinases (MMPs) are important for many different types of cancer-related processes, including metastasis. Understanding the functional impact of changes in MMP activity during cancer treatment is an important facet not typically evaluated as part of preclinical research. With MMP activity being a critical component of the metastatic cascade, we designed a 3D hydrogel system to probe whether pharmacological inhibition affected human melanoma cell proteolytic activity; metastatic melanoma is a highly aggressive and drug-resistant form of skin cancer. The relationship between MMP activity and drug treatment is unknown, and therefore we used an in situ fluorogenic MMP sensor peptide to determine how drug treatment affects melanoma cell MMP activity in three dimensions. We encapsulated melanoma cells from varying stages of progression within PEG-based hydrogels to examine the relationship between drug treatment and MMP activity. From these results, a metastatic melanoma cell line (A375) and two inhibitors that inhibit RAF (PLX4032 and sorafenib) were studied further to determine whether changes in MMP activity led to a functional change in cell behavior. A375 cells exhibited increased MMP activity despite an overall decrease in metabolic activity with PLX4032 treatment. The changes in proteolytic activity correlated with increased cell elongation and increased single-cell migration. In contrast, sorafenib did not alter MMP activity or cell motility, showing that the changes induced by PLX4032 were not a universal response to small-molecule inhibition. Therefore, we argue the importance of studying MMP activity with drug treatment and its possible implications for unwanted side effects.
Collapse
|
36
|
Harkness LM, Ashton AW, Burgess JK. Asthma is not only an airway disease, but also a vascular disease. Pharmacol Ther 2014; 148:17-33. [PMID: 25460035 DOI: 10.1016/j.pharmthera.2014.11.010] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 07/29/2014] [Indexed: 12/24/2022]
Abstract
Multiple studies have identified an expansion and morphological dysregulation of the bronchial vascular network in the airways of asthmatics. Increased number, size and density of blood vessels, as well as vascular leakage and plasma engorgement, have been reported in the airways of patients with all grades of asthma from mild to fatal. This neovascularisation is an increasingly commonly reported feature of airway remodelling; however, the pathophysiological impact of the increased vasculature in the bronchial wall and its significance to pulmonary function in asthma are unrecognised at this time. Multiple factors capable of influencing the development and persistence of the vascular network exist within asthmatic airway tissue. These include structural components of the altered extracellular matrix (ECM), imbalance of proteases and their endogenous inhibitors, release of active matrikines and the dysregulated levels of both soluble and matrix sequestered growth factors. This review will explore the features of the asthmatic airway which influence the development and persistence of the increased vascular network, as well as the effect of enhanced tissue perfusion on chronic inflammation and airway dynamics. The response of cells of the airways to the altered vascular profile and the subsequent influence on the features of airway remodelling will also be highlighted. We will explore the failure of current asthma therapeutics in "normalising" this vascular remodelling. Finally, we will summarize the outcomes of recent clinical trials which provide hope that anti-angiogenic therapies may be a potent asthma-resolving class of drugs and provide a new approach to asthma management in the future.
Collapse
Affiliation(s)
- Louise M Harkness
- Respiratory Cellular and Molecular Biology, Woolcock Institute of Medical Research, The University of Sydney, Sydney, NSW, Australia; Discipline of Pharmacology, The University of Sydney, Sydney, NSW, Australia
| | - Anthony W Ashton
- Division of Perinatal Research, Kolling Institute, Sydney, NSW, Australia
| | - Janette K Burgess
- Respiratory Cellular and Molecular Biology, Woolcock Institute of Medical Research, The University of Sydney, Sydney, NSW, Australia; Discipline of Pharmacology, The University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
37
|
Matusiak N, Castelli R, Tuin AW, Overkleeft HS, Wisastra R, Dekker FJ, Prély LM, Bischoff R, Bischoff RPM, van Waarde A, Dierckx RAJO, Elsinga PH. A dual inhibitor of matrix metalloproteinases and a disintegrin and metalloproteinases, [¹⁸F]FB-ML5, as a molecular probe for non-invasive MMP/ADAM-targeted imaging. Bioorg Med Chem 2014; 23:192-202. [PMID: 25438884 DOI: 10.1016/j.bmc.2014.11.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 11/06/2014] [Accepted: 11/06/2014] [Indexed: 11/17/2022]
Abstract
BACKGROUND Numerous clinical studies have shown a correlation between increased matrix metalloproteinase (MMP)/a disintegrin and metalloproteinase (ADAM) activity and poor outcome of cancer. Various MMP inhibitors (MMPIs) have been developed for therapeutic purposes in oncology. In addition, molecular imaging of MMP/ADAM levels in vivo would allow the diagnosis of tumors. We selected the dual inhibitor of MMPs and ADAMs, ML5, which is a hydroxamate-based inhibitor with affinities for many MMPs and ADAMs. ML5 was radiolabelled with (18)F and the newly obtained radiolabelled inhibitor was evaluated in vitro and in vivo. MATERIALS AND METHODS ML5 was radiolabelled by direct acylation with N-succinimidyl-4-[(18)F]fluorobenzoate ([(18)F]SFB) for PET (positron emission tomography). The resulting radiotracer [(18)F]FB-ML5 was evaluated in vitro in human bronchial epithelium 16HBE cells and breast cancer MCF-7 cells. The non-radioactive probe FB-ML5 and native ML5 were tested in a fluorogenic inhibition assay against MMP-2, -9, -12 and ADAM-17. The in vivo kinetics of [(18)F]FB-ML5 were examined in a HT1080 tumor-bearing mouse model. Specificity of probe binding was examined by co-injection of 0 or 2.5mg/kg ML5. RESULTS ML5 and FB-ML5 showed high affinity for MMP-2, -9, -12 and ADAM-17; indeed IC50 values were respectively 7.4 ± 2.0, 19.5 ± 2.8, 2.0 ± 0.2 and 5.7 ± 2.2 nM and 12.5 ± 3.1, 31.5 ± 13.7, 138.0 ± 10.9 and 24.7 ± 2.8 nM. Radiochemical yield of HPLC-purified [(18)F]FB-ML5 was 13-16% (corrected for decay). Cellular binding of [(18)F]FB-ML5 was reduced by 36.6% and 27.5% in MCF-7 and 16 HBE cells, respectively, after co-incubation with 10 μM of ML5. In microPET scans, HT1080 tumors exhibited a low and homogeneous uptake of the tracer. Tumors of mice injected with [(18)F]FB-ML5 showed a SUVmean of 0.145 ± 0.064 (n=6) which decreased to 0.041 ± 0.027 (n=6) after target blocking (p<0.05). Ex vivo biodistribution showed a rapid excretion through the kidneys and the liver. Metabolite assays indicated that the parent tracer represented 23.2 ± 7.3% (n=2) of total radioactivity in plasma, at 90 min post injection (p.i.). CONCLUSION The nanomolar affinity MMP/ADAM inhibitor ML5 was successfully labelled with (18)F. [(18)F]FB-ML5 demonstrated rather low binding in ADAM-17 overexpressing cell lines. [(18)F]FB-ML5 uptake showed significant reduction in the HT1080 tumor in vivo after co-injection of ML5. [(18)F]FB-ML5 may be suitable for the visualization/quantification of diseases overexpressing simultaneously MMPs and ADAMs.
Collapse
Affiliation(s)
- Nathalie Matusiak
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9700 RB Groningen, The Netherlands
| | - Riccardo Castelli
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Adriaan W Tuin
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | | | - Rosalina Wisastra
- Department of Pharmaceutical Gene Modulation, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Frank J Dekker
- Department of Pharmaceutical Gene Modulation, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Laurette M Prély
- Department of Analytical Biochemistry, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| | | | - Rainer P M Bischoff
- Department of Analytical Biochemistry, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Aren van Waarde
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9700 RB Groningen, The Netherlands
| | - Rudi A J O Dierckx
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9700 RB Groningen, The Netherlands
| | - Philip H Elsinga
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9700 RB Groningen, The Netherlands.
| |
Collapse
|
38
|
Grotz TE, Kottschade L, Pavey ES, Markovic SN, Jakub JW. Adjuvant GM-CSF improves survival in high-risk stage iiic melanoma: a single-center Study. Am J Clin Oncol 2014; 37:467-472. [PMID: 23428946 PMCID: PMC3664256 DOI: 10.1097/coc.0b013e31827def82] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
OBJECTIVES Stage III melanoma is associated with an increased risk of recurrence and death. Complete surgical resection remains the best chance for cure. Unfortunately, no adjuvant therapy has demonstrated a consistent improvement in melanoma-specific survival (MSS). We hypothesize that adjuvant granulocyte-macrophage colony-stimulating factor (GM-CSF) may improve clinical outcomes. PATIENTS AND METHODS Retrospective cohort study of 317 surgically resected stage III melanoma patients managed with observation or adjuvant GM-CSF at a single institution from 2001 to 2010. RESULTS Of the 317 stage III patients, 165 (52%) were observed and 152 (48%) were treated with GM-CSF, with a median follow-up of 34 months. Patients treated with GM-CSF tended to be younger (P < 0.0001), had more advanced stage disease (P = 0.002), and were more likely to have had a recurrence before initiation of adjuvant therapy than the observation group (P < 0.0001). Adjuvant GM-CSF seemed to be associated with improved MSS, but this did not reach statistical significance (P = 0.08). Patients with stage IIIC melanoma derived a substantial benefit from adjuvant GM-CSF, with a 52% risk reduction in melanoma-specific death (hazard ratio 0.48; 95% confidence interval, 0.27-0.87; P = 0.02). CONCLUSIONS Despite selecting patients with more advanced stage and a higher incidence of regional relapse, adjuvant GM-CSF was associated with an improved MSS but not disease-free survival in patients with stage IIIC disease. In patients not otherwise eligible for clinical trials, adjuvant GM-CSF treatment is a reasonable option for individuals with resected high-risk melanoma.
Collapse
Affiliation(s)
| | - Lisa Kottschade
- Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester MN
| | - Emily S. Pavey
- Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester MN
| | | | | |
Collapse
|
39
|
Verma S, Kesh K, Ganguly N, Jana S, Swarnakar S. Matrix metalloproteinases and gastrointestinal cancers: Impacts of dietary antioxidants. World J Biol Chem 2014; 5:355-376. [PMID: 25225603 PMCID: PMC4160529 DOI: 10.4331/wjbc.v5.i3.355] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 05/07/2014] [Accepted: 06/11/2014] [Indexed: 02/05/2023] Open
Abstract
The process of carcinogenesis is tightly regulated by antioxidant enzymes and matrix degrading enzymes, namely, matrix metalloproteinases (MMPs). Degradation of extracellular matrix (ECM) proteins like collagen, proteoglycan, laminin, elastin and fibronectin is considered to be the prerequisite for tumor invasion and metastasis. MMPs can degrade essentially all of the ECM components and, most MMPs also substantially contribute to angiogenesis, differentiation, proliferation and apoptosis. Hence, MMPs are important regulators of tumor growth both at the primary site and in distant metastases; thus the enzymes are considered as important targets for cancer therapy. The implications of MMPs in cancers are no longer mysterious; however, the mechanism of action is yet to be explained. Herein, our major interest is to clarify how MMPs are tied up with gastrointestinal cancers. Gastrointestinal cancer is a variety of cancer types, including the cancers of gastrointestinal tract and organs, i.e., esophagus, stomach, biliary system, pancreas, small intestine, large intestine, rectum and anus. The activity of MMPs is regulated by its endogenous inhibitor tissue inhibitor of metalloproteinase (TIMP) which bind MMPs with a 1:1 stoichiometry. In addition, RECK (reversion including cysteine-rich protein with kazal motifs) is a membrane bound glycoprotein that inhibits MMP-2, -9 and -14. Moreover, α2-macroglobulin mediates the uptake of several MMPs thereby inhibit their activity. Cancerous conditions increase intrinsic reactive oxygen species (ROS) through mitochondrial dysfunction leading to altered protease/anti-protease balance. ROS, an index of oxidative stress is also involved in tumorigenesis by activation of different MAP kinase pathways including MMP induction. Oxidative stress is involved in cancer by changing the activity and expression of regulatory proteins especially MMPs. Epidemiological studies have shown that high intake of fruits that rich in antioxidants is associated with a lower cancer incidence. Evidence indicates that some antioxidants inhibit the growth of malignant cells by inducing apoptosis and inhibiting the activity of MMPs. This review is discussed in six subchapters, as follows.
Collapse
|
40
|
Kavarana MN, Jones JA, Stroud RE, Bradley SM, Ikonomidis JS, Mukherjee R. Pulmonary arteriovenous malformations after the superior cavopulmonary shunt: mechanisms and clinical implications. Expert Rev Cardiovasc Ther 2014; 12:703-13. [PMID: 24758411 DOI: 10.1586/14779072.2014.912132] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Children with functional single ventricle heart disease are commonly palliated down a staged clinical pathway toward a Fontan completion procedure (total cavopulmonary connection). The Fontan physiology is fraught with long-term complications associated with lower body systemic venous hypertension, eventually resulting in significant morbidity and mortality. The bidirectional Glenn shunt or superior cavopulmonary connection (SCPC) is commonly the transitional stage in single ventricle surgical management and provides excellent palliation. Some studies have demonstrated lower morbidity and mortality with the SCPC when compared with the Fontan. Unfortunately the durability of the SCPC is significantly limited by the development of pulmonary arteriovenous malformations (PAVMs) which have been commonly attributed to the absence of hepatic venous blood flow and the lack of pulsatile flow to the affected lungs. Abnormal angiogenesis has been suggested as a final common pathway to PAVM development. Understanding these fundamental mechanisms through the investigation of angiogenic pathways associated with the pathogenesis of PAVMs would help to develop medical therapies that could prevent or reverse this complication following SCPC. Such therapies could improve the longevity of the SCPC, potentially eliminate or significantly postpone the Fontan completion with its associated complications, and improve long-term survival in children with single ventricle disease.
Collapse
Affiliation(s)
- Minoo N Kavarana
- Section of Pediatric Cardiothoracic Surgery, Department of Surgery, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC 29425, USA
| | | | | | | | | | | |
Collapse
|
41
|
de Vega S, Suzuki N, Nonaka R, Sasaki T, Forcinito P, Arikawa-Hirasawa E, Yamada Y. A C-terminal fragment of fibulin-7 interacts with endothelial cells and inhibits their tube formation in culture. Arch Biochem Biophys 2014; 545:148-53. [PMID: 24480309 PMCID: PMC3974681 DOI: 10.1016/j.abb.2014.01.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 12/21/2013] [Accepted: 01/15/2014] [Indexed: 11/17/2022]
Abstract
We have previously demonstrated that fibulin-7 (Fbln7) is expressed in teeth by pre-odontoblast and odontoblast cells, localized in the basement membrane and dentin matrices, and is an adhesion molecule for dental mesenchyme cells and odontoblasts. Fbln7 is also expressed in blood vessels by endothelial cells. In this report, we show that a recombinant C-terminal Fbln7 fragment (Fbln7-C) bound to Human Umbilical Vein Endothelial Cells (HUVECs) but did not promote cell spreading and actin stress fiber formation. Fbln7-C binding to HUVECs induced integrin clustering at cell adhesion sites with other focal adhesion molecules, and sustained activation of FAK, p130Cas, and Rac1. In addition, RhoA activation was inhibited, thereby preventing HUVEC spreading. As endothelial cell spreading is an important step for angiogenesis, we examined the effect of Fbln7-C on angiogenesis using in vitro assays for endothelial cell tube formation and vessel sprouting from aortic rings. We found that Fbln7-C inhibited the HUVEC tube formation and the vessel sprouting in aortic ring assays. Our findings suggest potential anti-angiogenic activity of the Fbln7 C-terminal region.
Collapse
Affiliation(s)
- Susana de Vega
- Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892-4370, United States; Research Institute for Diseases of Old Age, Juntendo University School of Medicine, Tokyo 113-8421, Japan
| | - Nobuharu Suzuki
- Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892-4370, United States; Department of Biochemistry and Biophysics, Graduate School of Health Care Sciences, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Risa Nonaka
- Research Institute for Diseases of Old Age, Juntendo University School of Medicine, Tokyo 113-8421, Japan
| | - Takako Sasaki
- Department of Experimental Medicine, Nikolaus Fiebiger Center of Molecular Medicine, University of Erlangen-Nuernberg, Erlangen, Germany
| | - Patricia Forcinito
- Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892-4370, United States
| | - Eri Arikawa-Hirasawa
- Research Institute for Diseases of Old Age, Juntendo University School of Medicine, Tokyo 113-8421, Japan
| | - Yoshihiko Yamada
- Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892-4370, United States.
| |
Collapse
|
42
|
Breast carcinoma progression and tumour vascular markers related to apoptotic mechanisms. DISEASE MARKERS 2014; 2014:156034. [PMID: 24696529 PMCID: PMC3948469 DOI: 10.1155/2014/156034] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 01/03/2014] [Indexed: 02/06/2023]
Abstract
Background. In the last few years, the cancer research had tried to identify and characterize new biochemical and molecular pathways in which the inhibition induces prosurvival mechanisms. Our work describes the expression of two different members of apoptotic regulatory pathway and their relationship with a progression of breast carcinoma. Materials and Methods. We compared expression of genes related to apoptosis (DR6 and Gpm6B) in the blood of patients suffering from stage I of breast cancer in different grades (I–IV), with healthy controls. After isolation of mRNA, transcription of mRNA into the cDNA was performed. The quantification of gene expression changes in DR6 and Gpm6B was detected by RT-PCR method. Analysis at the protein level was performed by the Western blot.Results. In statistical analysis of Dr6 mRNA level changes we detected significant increase starting in Grading 1 (G1) and reached maximal level in G3.This expression on mRNA levels was similar to protein levels, which copy rising tendency with maximal value in G3. The results of Gpm6B were significantly lower.Conclusion. This result showed that antiapoptotic signalling during neovascularization is increased significantly. It would be advisable in the future to study the influence of cytostatic treatment on the expression of genes related to apoptotic pathways and their relationship with progression of breast cancer tumours.
Collapse
|
43
|
van Tilborg AAG, Sweep FCGJ, Geurts-Moespot AJ, Wetzels AMM, de Waal RMW, Westphal JR, Massuger LFAG. Plasminogen activators are involved in angiostatin generation in vivo in benign and malignant ovarian tumor cyst fluids. Int J Oncol 2014; 44:1394-400. [PMID: 24535412 DOI: 10.3892/ijo.2014.2303] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Accepted: 01/22/2014] [Indexed: 11/05/2022] Open
Abstract
In many tumor types, angiogenesis is the net result of pro- and anti-angiogenic mediators and correlated with metabolic activity, growth, and degree of malignancy. One of the first discovered anti-angiogenic compounds is angiostatin, a proteolytic fragment of plasminogen. The requirements for in vivo angiostatin generation have not yet been determined. We investigated the levels of plasminogen and angiostatin by western blotting and of components of the plasminogen activator complex by ELISA in cyst fluid derived from benign and malignant ovarian tumors. Fluid samples from functional ovarian follicles, dermoid cysts and endometriotic lesions were evaluated separately. When no or minimal amounts of plasminogen were present in the cyst fluids, angiostatin was generally absent as well, irrespective of plasminogen activator concentrations. When plasminogen was present, the degree of conversion of plasminogen to angiostatin was significantly correlated with the level of uPA, and, to a lesser extent, to the tPA level. However, angiostatin was also found in a number of cyst fluid samples with minimal or no plasminogen activators, suggesting the involvement of other angiostatin generating proteases in these samples. Conversely, no angiostatin was observed in a number of cyst fluid samples containing both plasminogen and plasminogen activators. The presence of an inhibitor of the enzymatic activity of uPA and/or tPA, like PAI-1, may explain this finding. Our data show that plasminogen activators are clearly involved in in vivo angiostatin formation in ovarian cysts. Most likely, however, other proteases, as well as inhibitors of plasminogen activators, are involved as well.
Collapse
Affiliation(s)
- A A G van Tilborg
- Department of Obstetrics and Gynaecology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - F C G J Sweep
- Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - A J Geurts-Moespot
- Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - A M M Wetzels
- Department of Obstetrics and Gynaecology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - R M W de Waal
- Department of Obstetrics and Gynaecology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - J R Westphal
- Department of Obstetrics and Gynaecology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - L F A G Massuger
- Department of Obstetrics and Gynaecology, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
44
|
Matrix metalloproteinases: the gene expression signatures of head and neck cancer progression. Cancers (Basel) 2014; 6:396-415. [PMID: 24531055 PMCID: PMC3980592 DOI: 10.3390/cancers6010396] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 01/14/2014] [Accepted: 01/29/2014] [Indexed: 11/17/2022] Open
Abstract
Extracellular matrix degradation by matrix metalloproteinases (MMPs) plays a pivotal role in cancer progression by promoting motility, invasion and angiogenesis. Studies have shown that MMP expression is increased in head and neck squamous cell carcinomas (HNSCCs), one of the most common cancers in the world, and contributes to poor outcome. In this review, we examine the expression pattern of MMPs in HNSCC by microarray datasets and summarize the current knowledge of MMPs, specifically MMP-1, -3, -7 -10, -12, -13, 14 and -19, that are highly expressed in HNSCCs and involved cancer invasion and angiogenesis.
Collapse
|
45
|
Dodd T, Wiggins L, Hutcheson R, Smith E, Musiyenko A, Hysell B, Russell JC, Rocic P. Impaired coronary collateral growth in the metabolic syndrome is in part mediated by matrix metalloproteinase 12-dependent production of endostatin and angiostatin. Arterioscler Thromb Vasc Biol 2013; 33:1339-49. [PMID: 23599440 DOI: 10.1161/atvbaha.113.301533] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE We have previously shown that transient coronary artery occlusion stimulated coronary collateral growth (CCG) in healthy (Sprague Dawley) but not in metabolic syndrome (JCR:LA-cp [JCR] ) rats. Here, we sought to determine whether matrix metalloproteinases (MMPs) negatively regulate CCG in the metabolic syndrome via release of endostatin and angiostatin. APPROACH AND RESULTS Rats underwent transient, repetitive left anterior descending occlusion and resultant myocardial ischemia (RI) for 0 to 10 days. CCG was measured in the collateral-dependent and normal zones using microspheres, MMP activation by Western blot, and endostatin and angiostatin by ELISA on days 0, 3, 6, 9, or 10 of RI. Endostatin and angiostatin were increased in JCR but not in Sprague Dawley rats on days 6 and 9 of RI. Increased endostatin and angiostatin correlated with increased MMP12 (≈ 4-fold) activation in JCR but not in Sprague Dawley rats on days 6 and 9 of RI. Inhibition of MMP12 in JCR rats nearly completely blocked endostatin (≈ 85%) and angiostatin (≈ 90%) generation and significantly improved CCG (collateral-dependent zone flow was ≈ 66% of normal zone flow versus ≈ 12% for JCR RI). CONCLUSIONS Compromised CCG in the metabolic syndrome is, in large part, because of increased MMP12 activation and consequent increased generation of endostatin and angiostatin, which inhibits late-stage collateral remodeling.
Collapse
Affiliation(s)
- Tracy Dodd
- Department of Biochemistry and Molecular Biology, University of South Alabama College of Medicine, Mobile, AL 36688, USA
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Missing the target: matrix metalloproteinase antitargets in inflammation and cancer. Trends Pharmacol Sci 2013; 34:233-42. [PMID: 23541335 DOI: 10.1016/j.tips.2013.02.004] [Citation(s) in RCA: 278] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 02/15/2013] [Accepted: 02/19/2013] [Indexed: 01/08/2023]
Abstract
Matrix metalloproteinases (MMPs) are reputed to cause the inflammatory tissue destruction characterizing chronic inflammatory diseases and to degrade basement membrane collagen, thereby facilitating cancer cell metastasis. However, following the disappointing MMP drug cancer trials, recent studies using mouse models of disease coupled with high-throughput methods for substrate discovery have revealed surprising and unexpected new biological roles of MMPs in inflammatory diseases and cancer in vivo. Thus, MMPs modify signaling pathways and regulate the activity of whole families of cytokines of the immune response by precise proteolytic processing. By cleaving and inactivating cytokine-binding proteins and protease inhibitors, cytokine activities are unmasked and activities of diverse proteases are increased in an interconnected protease web. With new substrates come new roles, and 10 of 24 murine MMPs have antitumorigenic and anti-inflammatory roles making them drug antitargets; that is, their beneficial actions should not be inhibited. Here, we examine whether the discovery that MMPs are drug antitargets for one disease might pave the way for their use for other indications or whether this is a serious threat to the development of MMP inhibitors.
Collapse
|
47
|
Vihinen P, Ala-Aho R, Kähäri VM. Diagnostic and prognostic role of matrix metalloproteases in cancer. ACTA ACUST UNITED AC 2013; 2:1025-39. [PMID: 23495924 DOI: 10.1517/17530059.2.9.1025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Matrix metalloproteases (MMPs) are key players in the progression and metastasis of cancer. MMPs cleave extracellular matrix components and in this way promote tumor growth, invasion and vascularization. MMPs also affect tumor progression by regulating availability and activity of growth factors, inflammatory cytokines and chemokines. Accordingly, several MMPs have been found to serve as prognostic indicators in solid tumors. Usually the increased levels of MMPs in patients' tumor tissue or serum/plasma are associated with poor outcome. Interestingly, recent results show that certain MMPs also serve as tumor suppressors. OBJECTIVE This review discusses the latest view on MMPs as diagnostic and prognostic indicators in cancer patients. METHODS Studies with clinical samples of 70 or more patients are included in particular. In addition, the possible roles of MMPs in future molecular diagnostics and in the evaluation of therapeutic responses are discussed. CONCLUSION MMP-9 in particular has shown prognostic value in various types of tumor, and its measurement in circulation, urine or tumor tissue might help in clinical surveillance of otherwise problematic patient cases. There is upcoming new knowledge on MMPs in therapy response evaluation, in which MMPs might be useful together with CT scans and other clinically more established prognostic factors. Certain MMPs have a dual role in terms of cancer-modulating properties and thus it is essential to evaluate their expression and function in tumor cells and host environment to select validated therapy targets but spare MMP antitargets.
Collapse
Affiliation(s)
- Pia Vihinen
- Turku University Hospital, Department of Oncology and Radiotherapy, POB 52, FIN-20521 Turku, Finland +358 2 313 0804 ; +358 2 313 2809 ;
| | | | | |
Collapse
|
48
|
Watnick RS. The role of the tumor microenvironment in regulating angiogenesis. Cold Spring Harb Perspect Med 2012; 2:a006676. [PMID: 23209177 DOI: 10.1101/cshperspect.a006676] [Citation(s) in RCA: 111] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The tumor-associated stroma has been shown to play a significant role in cancer formation. Paracrine signaling interactions between epithelial tumor cells and stromal cells are a key component in the transformation and proliferation of tumors in several organs. Whereas the intracellular signaling pathways regulating the expression of several pro- and antiangiogenic proteins have been well characterized in human cancer cells, the intercellular signaling that takes place between tumor cells and the surrounding tumor-associated stroma has not been as extensively studied with regard to the regulation of angiogenesis. In this chapter we define the key players in the regulation of angiogenesis and examine how their expression is regulated in the tumor-associated stroma. The resulting analysis is often seemingly paradoxical, underscoring the complexity of intercellular signaling within tumors and the need to better understand the environmental context underlying these signaling mechanisms.
Collapse
Affiliation(s)
- Randolph S Watnick
- Children's Hospital Boston, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
49
|
Morrison BJ, Steel JC, Morris JC. Sphere culture of murine lung cancer cell lines are enriched with cancer initiating cells. PLoS One 2012; 7:e49752. [PMID: 23152931 PMCID: PMC3496706 DOI: 10.1371/journal.pone.0049752] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Accepted: 10/15/2012] [Indexed: 02/06/2023] Open
Abstract
Cancer initiating cells (CICs) represent a unique cell population essential for the maintenance and growth of tumors. Most in vivo studies of CICs utilize human tumor xenografts in immunodeficient mice. These models provide limited information on the interaction of CICs with the host immune system and are of limited value in assessing therapies targeting CICs, especially immune-based therapies. To assess this, a syngeneic cancer model is needed. We examined the sphere-forming capacity of thirteen murine lung cancer cell lines and identified TC-1 and a metastatic subclone of Lewis lung carcinoma (HM-LLC) as cell lines that readily formed and maintained spheres over multiple passages. TC-1 tumorspheres were not enriched for expression of CD133 or CD44, putative CIC markers, nor did they demonstrate Hoechst 33342 side population staining or Aldefluor activity compared to adherent TC-1 cells. However, in tumorsphere culture, these cells exhibited self-renewal and long-term symmetric division capacity and expressed more Oct-4 compared to adherent cells. HM-LLC sphere-derived cells exhibited increased Oct-4, CD133, and CD44 expression, demonstrated a Hoechst 33342 side population and Aldefluor activity compared to adherent cells or a low metastatic subclone of LLC (LM-LLC). In syngeneic mice, HM-LLC sphere-derived cells required fewer cells to initiate tumorigenesis compared to adherent or LM-LLC cells. Similarly TC-1 sphere-derived cells were more tumorigenic than adherent cells in syngeneic mice. In contrast, in immunocompromised mice, less than 500 sphere or adherent TC-1 cells and less than 1,000 sphere or adherent LLC cells were required to initiate a tumor. We suggest that no single phenotypic marker can identify CICs in murine lung cancer cell lines. Tumorsphere culture may provide an alternative approach to identify and enrich for murine lung CICs. Furthermore, we propose that assessing tumorigenicity of murine lung CICs in syngeneic mice better models the interaction of CICs with the host immune system.
Collapse
MESH Headings
- AC133 Antigen
- Animals
- Antigens, CD/metabolism
- Benzimidazoles/metabolism
- Carcinoma, Lewis Lung/genetics
- Carcinoma, Lewis Lung/pathology
- Cell Adhesion
- Cell Membrane/metabolism
- Cell Transformation, Neoplastic/pathology
- Clone Cells
- Gene Expression Regulation, Neoplastic
- Glycoproteins/metabolism
- Humans
- Hyaluronan Receptors/metabolism
- Immunocompetence
- Immunocompromised Host
- Lung Neoplasms/genetics
- Lung Neoplasms/pathology
- Mice
- Mice, Inbred C57BL
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Octamer Transcription Factor-3/genetics
- Octamer Transcription Factor-3/metabolism
- Peptides/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Spheroids, Cellular/metabolism
- Spheroids, Cellular/pathology
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Brian J. Morrison
- Division of Hematology-Oncology, Department of Medicine, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Jason C. Steel
- Division of Hematology-Oncology, Department of Medicine, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - John C. Morris
- Division of Hematology-Oncology, Department of Medicine, University of Cincinnati, Cincinnati, Ohio, United States of America
- * E-mail:
| |
Collapse
|
50
|
Radziwon-Balicka A, Moncada de la Rosa C, Jurasz P. Platelet-associated angiogenesis regulating factors: a pharmacological perspective. Can J Physiol Pharmacol 2012; 90:679-88. [DOI: 10.1139/y2012-036] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Platelets, in addition to maintaining hemostasis, also stimulate angiogenesis by generating and releasing, upon activation, factors that promote the growth of new blood vessels. To date, at least 20 angiogenesis-regulating factors have been identified in platelets, including both promoters and inhibitors. Platelet-derived angiogenesis regulators promote angiogenesis during wound healing, tumor growth, and in response to ischemia. Within platelets, angiogenesis regulators are primarily stored in α-granules, but are also found in the cytosol or derived from membrane lipids. Their release can be inhibited pharmacologically by anti-platelet agents, which consequently suppress platelet-stimulated angiogenesis. Several years ago, our research group discovered that platelets generate the angiogenesis inhibitor angiostatin independent of the activation state of platelets, and that platelet-derived angiostatin serves to limit the angiogenesis-stimulating effects of platelets. In this review, we summarize the current knowledge of platelet-associated angiogenesis regulators, how they impact angiogenesis, and how they are controlled pharmacologically.
Collapse
Affiliation(s)
- Aneta Radziwon-Balicka
- Faculty of Pharmacy and Pharmaceutical Sciences, 3-142E Katz Group-Rexall Centre for Pharmacy & Health Research, University of Alberta, Edmonton, AB T6G 2E1, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, AB T6G 2S2, Canada
| | - Cesar Moncada de la Rosa
- Faculty of Pharmacy and Pharmaceutical Sciences, 3-142E Katz Group-Rexall Centre for Pharmacy & Health Research, University of Alberta, Edmonton, AB T6G 2E1, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, AB T6G 2S2, Canada
| | - Paul Jurasz
- Faculty of Pharmacy and Pharmaceutical Sciences, 3-142E Katz Group-Rexall Centre for Pharmacy & Health Research, University of Alberta, Edmonton, AB T6G 2E1, Canada
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, AB T6G 2S2, Canada
| |
Collapse
|