51
|
High MMP14 expression is predictive of poor prognosis in resectable hepatocellular carcinoma. Pathology 2020; 52:359-365. [PMID: 32122646 DOI: 10.1016/j.pathol.2020.01.436] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 01/08/2020] [Accepted: 01/20/2020] [Indexed: 12/18/2022]
Abstract
Matrix metalloproteinase 14 (MMP14) has been found to play multiple biological roles in cancers, including hepatocellular carcinoma (HCC). Up to now, its expression, clinicopathological and prognostic implications in HCC have not been comprehensively investigated. In the present study, MMP14 expression was detected, using tissue microarray-based immunohistochemical staining, in paired HCC and adjacent liver (AL) samples from 260 patients who underwent radical hepatectomy. The associations of MMP14 staining H-scores with clinicopathological parameters, overall and disease-free survival were then evaluated. Finally, its expression and prognostic value were confirmed in some online publicly available databases. It was shown that MMP14 expression was significantly higher in HCC than in AL tissues (p=0.035). Furthermore, MMP14 expression correlated positively with tumour size, Edmondson-Steiner grade and α-fetoprotein level (p<0.05). For survival, MMP14 expression was negatively associated with both overall and disease-free survival in univariate analyses (p<0.05), while it remained statistically significant for disease-free survival by multivariate Cox regression test. In the Ualcan and Kaplan-Meier Plotter databases, MMP14 was also revealed to be overexpressed and prognostic. Taken together, our study indicated that high MMP14 expression was predictive for unfavourable biological behaviours and long-term prognosis in resectable HCC.
Collapse
|
52
|
Kusiak AA, Szopa MD, Jakubowska MA, Ferdek PE. Signaling in the Physiology and Pathophysiology of Pancreatic Stellate Cells - a Brief Review of Recent Advances. Front Physiol 2020; 11:78. [PMID: 32116785 PMCID: PMC7033654 DOI: 10.3389/fphys.2020.00078] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 01/23/2020] [Indexed: 12/12/2022] Open
Abstract
The interest in pancreatic stellate cells (PSCs) has been steadily growing over the past two decades due mainly to the central role these cells have in the desmoplastic reaction associated with diseases of the pancreas, such as pancreatitis or pancreatic cancer. In recent years, the scientific community has devoted substantial efforts to understanding the signaling pathways that govern PSC activation and interactions with neoplastic cells. This mini review aims to summarize some very recent findings on signaling in PSCs and highlight their impact to the field.
Collapse
Affiliation(s)
- Agnieszka A Kusiak
- Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Mateusz D Szopa
- Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | | | - Pawel E Ferdek
- Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| |
Collapse
|
53
|
Dosch AR, Dai X, Reyzer ML, Mehra S, Srinivasan S, Willobee BA, Kwon D, Kashikar N, Caprioli R, Merchant NB, Nagathihalli NS. Combined Src/EGFR Inhibition Targets STAT3 Signaling and Induces Stromal Remodeling to Improve Survival in Pancreatic Cancer. Mol Cancer Res 2020; 18:623-631. [PMID: 31949002 DOI: 10.1158/1541-7786.mcr-19-0741] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 11/26/2019] [Accepted: 01/10/2020] [Indexed: 02/07/2023]
Abstract
Lack of durable response to cytotoxic chemotherapy is a major contributor to the dismal outcomes seen in pancreatic ductal adenocarcinoma (PDAC). Extensive tumor desmoplasia and poor vascular supply are two predominant characteristics which hinder the delivery of chemotherapeutic drugs into PDAC tumors and mediate resistance to therapy. Previously, we have shown that STAT3 is a key biomarker of therapeutic resistance to gemcitabine treatment in PDAC, which can be overcome by combined inhibition of the Src and EGFR pathways. Although it is well-established that concurrent EGFR and Src inhibition exert these antineoplastic properties through direct inhibition of mitogenic pathways in tumor cells, the influence of this combined therapy on stromal constituents in PDAC tumors remains unknown. In this study, we demonstrate in both orthotopic tumor xenograft and Ptf1acre/+;LSL-KrasG12D/+;Tgfbr2flox/flox (PKT) mouse models that concurrent EGFR and Src inhibition abrogates STAT3 activation, increases microvessel density, and prevents tissue fibrosis in vivo. Furthermore, the stromal changes induced by parallel EGFR and Src pathway inhibition resulted in improved overall survival in PKT mice when combined with gemcitabine. As a phase I clinical trial utilizing concurrent EGFR and Src inhibition with gemcitabine has recently concluded, these data provide timely translational insight into the novel mechanism of action of this regimen and expand our understanding into the phenomenon of stromal-mediated therapeutic resistance. IMPLICATIONS: These findings demonstrate that Src/EGFR inhibition targets STAT3, remodels the tumor stroma, and results in enhanced delivery of gemcitabine to improve overall survival in a mouse model of PDAC.
Collapse
Affiliation(s)
- Austin R Dosch
- Division of Surgical Oncology, Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida.,Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| | - Xizi Dai
- Division of Surgical Oncology, Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida.,Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| | - Michelle L Reyzer
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, Tennessee
| | - Siddharth Mehra
- Division of Surgical Oncology, Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida.,Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| | - Supriya Srinivasan
- Division of Surgical Oncology, Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida.,Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| | - Brent A Willobee
- Division of Surgical Oncology, Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida.,Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| | - Deukwoo Kwon
- Department of Public Health, University of Miami Miller School of Medicine, Miami, Florida
| | - Nilesh Kashikar
- Department of Pathology, University of Colorado, Denver, Colorado
| | - Richard Caprioli
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, Tennessee
| | - Nipun B Merchant
- Division of Surgical Oncology, Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida. .,Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| | - Nagaraj S Nagathihalli
- Division of Surgical Oncology, Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida. .,Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| |
Collapse
|
54
|
Willumsen N, Ali SM, Leitzel K, Drabick JJ, Yee N, Polimera HV, Nagabhairu V, Krecko L, Ali A, Maddukuri A, Moku P, Ali A, Poulose J, Menon H, Pancholy N, Costa L, Karsdal MA, Lipton A. Collagen fragments quantified in serum as measures of desmoplasia associate with survival outcome in patients with advanced pancreatic cancer. Sci Rep 2019; 9:19761. [PMID: 31875000 PMCID: PMC6930304 DOI: 10.1038/s41598-019-56268-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 12/08/2019] [Indexed: 02/07/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) patients have poor prognosis and poor response to treatment. This is largely due to PDAC being associated with a dense and active stroma and tumor fibrosis (desmoplasia). Desmoplasia is characterized by excessive degradation and formation of the extracellular matrix (ECM) generating collagen fragments that are released into circulation. We evaluated the association of specific collagen fragments measured in pre-treatment serum with outcome in patients with PDAC. Matrix metalloprotease (MMP)-degraded type I collagen (C1M), type III collagen (C3M), type IV collagen (C4M) and a pro-peptide of type III collagen (PRO-C3) were measured by ELISA in pre-treatment serum from a randomized phase 3 clinical trial of patients with stage III/IV PDAC treated with 5-fluorouracil based therapy (n = 176). The collagen fragments were evaluated for their correlation (r, Spearman) with serum CA19-9 and for their association with overall survival (OS) based on Cox-regression analyses. In this phase 3 PDAC trial, pre-treatment serum collagen fragment levels were above the reference range for 67%-98% of patients, with median values in PDAC approximately two-fold higher than reference levels. Collagen fragment levels did not correlate with CA19-9 (r = 0.049–0.141, p = ns). On a continuous basis, higher levels of all collagen fragments were associated with significantly shorter OS. When evaluating degradation (C3M) and formation (PRO-C3) of type III collagen further, higher PRO-C3 was associated with poor OS (>25th percentile cut-point, HR = 2.01, 95%CI = 1.33–3.05) and higher C3M/PRO-C3 ratio was associated with improved OS (>25th percentile cut-point, HR = 0.53, 95%CI = 0.34–0.80). When adjusting for CA19–9 and clinical covariates, PRO-C3 remained significant (HR = 1.65, 95%CI = 1.09–2.48). In conclusion, collagen remodeling quantified in pre-treatment serum as a surrogate measure of desmoplasia was significantly associated with OS in a phase 3 clinical PDAC trial, supporting the link between desmoplasia, tumorigenesis, and response to treatment. If validated, these biomarkers may have prognostic and/or predictive potential in future PDAC trials.
Collapse
Affiliation(s)
| | - Suhail M Ali
- Division of Hematology/Oncology, Penn State Health Milton S Hershey Medical Center, Hershey, PA, USA.,Lebanon VA Medical Center, Lebanon, PA, USA
| | - Kim Leitzel
- Division of Hematology/Oncology, Penn State Health Milton S Hershey Medical Center, Hershey, PA, USA
| | - Joseph J Drabick
- Division of Hematology/Oncology, Penn State Health Milton S Hershey Medical Center, Hershey, PA, USA
| | - Nelson Yee
- Division of Hematology/Oncology, Penn State Health Milton S Hershey Medical Center, Hershey, PA, USA
| | - Hyma V Polimera
- Division of Hematology/Oncology, Penn State Health Milton S Hershey Medical Center, Hershey, PA, USA
| | - Vinod Nagabhairu
- Pinnacle Health System, University of Pittsburgh Medical Center, Harrisburg, PA, USA
| | - Laura Krecko
- Division of Hematology/Oncology, Penn State Health Milton S Hershey Medical Center, Hershey, PA, USA
| | - Ayesha Ali
- Division of Hematology/Oncology, Penn State Health Milton S Hershey Medical Center, Hershey, PA, USA
| | - Ashok Maddukuri
- Division of Hematology/Oncology, Penn State Health Milton S Hershey Medical Center, Hershey, PA, USA
| | - Prashanth Moku
- Division of Hematology/Oncology, Penn State Health Milton S Hershey Medical Center, Hershey, PA, USA
| | - Aamnah Ali
- Division of Hematology/Oncology, Penn State Health Milton S Hershey Medical Center, Hershey, PA, USA
| | - Joyson Poulose
- Division of Hematology/Oncology, Penn State Health Milton S Hershey Medical Center, Hershey, PA, USA
| | - Harry Menon
- Division of Hematology/Oncology, Penn State Health Milton S Hershey Medical Center, Hershey, PA, USA
| | - Neha Pancholy
- Division of Hematology/Oncology, Penn State Health Milton S Hershey Medical Center, Hershey, PA, USA
| | - Luis Costa
- Oncology division, Hospital de Santa Maria, Lisboa, Portugal.,Clinical Translational Oncology Research Unit, Institute of Molecular Medicine, Lisboa, Portugal
| | | | - Allan Lipton
- Division of Hematology/Oncology, Penn State Health Milton S Hershey Medical Center, Hershey, PA, USA
| |
Collapse
|
55
|
Roger E, Martel S, Bertrand-Chapel A, Depollier A, Chuvin N, Pommier RM, Yacoub K, Caligaris C, Cardot-Ruffino V, Chauvet V, Aires S, Mohkam K, Mabrut JY, Adham M, Fenouil T, Hervieu V, Broutier L, Castets M, Neuzillet C, Cassier PA, Tomasini R, Sentis S, Bartholin L. Schwann cells support oncogenic potential of pancreatic cancer cells through TGFβ signaling. Cell Death Dis 2019; 10:886. [PMID: 31767842 PMCID: PMC6877617 DOI: 10.1038/s41419-019-2116-x] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 10/09/2019] [Accepted: 11/05/2019] [Indexed: 02/06/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the solid tumors with the poorest prognosis. The stroma of this tumor is abundant and composed of extracellular matrix and stromal cells (including cancer-associated fibroblasts and immune cells). Nerve fibers invading this stroma represent a hallmark of PDAC, involved in neural remodeling, which participates in neuropathic pain, cancer cell dissemination and tumor relapse after surgery. Pancreatic cancer-associated neural remodeling is regulated through functional interplays mediated by physical and molecular interactions between cancer cells, nerve cells and surrounding Schwann cells, and other stromal cells. In the present study, we show that Schwann cells (glial cells supporting peripheral neurons) can enhance aggressiveness (migration, invasion, tumorigenicity) of pancreatic cancer cells in a transforming growth factor beta (TGFβ)-dependent manner. Indeed, we reveal that conditioned medium from Schwann cells contains high amounts of TGFβ able to activate the TGFβ-SMAD signaling pathway in cancer cells. We also observed in human PDAC samples that high levels of TGFβ signaling activation were positively correlated with perineural invasion. Secretome analyses by mass spectrometry of Schwann cells and pancreatic cancer cells cultured alone or in combination highlighted the central role of TGFβ in neuro-epithelial interactions, as illustrated by proteomic signatures related to cell adhesion and motility. Altogether, these results demonstrate that Schwann cells are a meaningful source of TGFβ in PDAC, which plays a crucial role in the acquisition of aggressive properties by pancreatic cancer cells.
Collapse
Affiliation(s)
- Elodie Roger
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon (CRCL), Lyon, 69373, France
| | - Sylvie Martel
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon (CRCL), Lyon, 69373, France
| | - Adrien Bertrand-Chapel
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon (CRCL), Lyon, 69373, France
| | - Arnaud Depollier
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon (CRCL), Lyon, 69373, France
| | - Nicolas Chuvin
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon (CRCL), Lyon, 69373, France.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Roxane M Pommier
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon (CRCL), Lyon, 69373, France
| | - Karam Yacoub
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon (CRCL), Lyon, 69373, France
| | - Cassandre Caligaris
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon (CRCL), Lyon, 69373, France
| | - Victoire Cardot-Ruffino
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon (CRCL), Lyon, 69373, France
| | - Véronique Chauvet
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon (CRCL), Lyon, 69373, France
| | - Sophie Aires
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon (CRCL), Lyon, 69373, France
| | - Kayvan Mohkam
- Hospices Civils de Lyon, Croix Rousse hospital, Claude-Bernard Lyon 1 University, Department of General Surgery & Liver Transplantation, Lyon, France
| | - Jean-Yves Mabrut
- Hospices Civils de Lyon, Croix Rousse hospital, Claude-Bernard Lyon 1 University, Department of General Surgery & Liver Transplantation, Lyon, France
| | - Mustapha Adham
- Hospices Civils de Lyon, Edouard Herriot hospital, Claude-Bernard Lyon 1 University, Department of General Surgery & Liver Transplantation, Lyon, France
| | - Tanguy Fenouil
- Hospices Civils de Lyon Institute of Pathology EST, CRCL INSERM U1052, University Lyon 1, Lyon, France
| | - Valérie Hervieu
- Hospices Civils de Lyon Institute of Pathology EST, CRCL INSERM U1052, University Lyon 1, Lyon, France
| | - Laura Broutier
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon (CRCL), Lyon, 69373, France
| | - Marie Castets
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon (CRCL), Lyon, 69373, France
| | - Cindy Neuzillet
- Medical Oncology Department, Curie Institute, Versailles Saint-Quentin University, 35 rue Dailly, 92210, Saint Cloud, France
| | - Philippe A Cassier
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon (CRCL), Lyon, 69373, France.,Departement d'Oncologie Médicale, Centre Léon Bérard, Lyon, 69008, France
| | - Richard Tomasini
- Aix-Marseille Université, Institut Paoli-Calmettes, INSERM U1068, CNRS UMR 7258, Centre de Recherche en Cancérologie de Marseille, Marseille, France
| | - Stéphanie Sentis
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon (CRCL), Lyon, 69373, France
| | - Laurent Bartholin
- Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon (CRCL), Lyon, 69373, France.
| |
Collapse
|
56
|
Aizawa T, Karasawa H, Funayama R, Shirota M, Suzuki T, Maeda S, Suzuki H, Yamamura A, Naitoh T, Nakayama K, Unno M. Cancer-associated fibroblasts secrete Wnt2 to promote cancer progression in colorectal cancer. Cancer Med 2019; 8:6370-6382. [PMID: 31468733 PMCID: PMC6797671 DOI: 10.1002/cam4.2523] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 08/12/2019] [Accepted: 08/15/2019] [Indexed: 12/11/2022] Open
Abstract
Recent studies have shown that the tumor microenvironment plays a significant role in the progression of solid tumors. As an abundant component of the tumor microenvironment, cancer-associated fibroblasts (CAFs) have been shown to promote tumorigenesis and cancer aggressiveness, but their molecular characteristics remain poorly understood. In the present study, paired CAFs and normal fibroblasts (NFs) were isolated from five colorectal cancer (CRC) tissues from patients who underwent surgical resection. The gene expression profiles of CAFs and NFs identified by RNA sequencing were compared to understand the complex role of CAFs in cancer progression. Gene Set Enrichment Analysis revealed that the gene sets related to the Wnt signaling pathway were highly enriched in CAFs, as well as TGFβ signaling, which is considered to be a regulator of CAFs. Among the components of this pathway, Wnt2 was specifically expressed. The observations led us to speculate that Wnt2 is extremely involved in regulating CRC progression by CAFs. Thus, we performed immunohistochemical analysis on Wnt2 in 171 patients who underwent surgery for colorectal adenocarcinoma. Positive staining for Wnt2 was mainly observed in cancer stroma, although the immunoreactivity was weak in cancer cells. Wnt2 expression in CAFs was significantly correlated with depth of tumor (P < .001), lymph node metastasis (P = .044), TNM stage (P = .010), venous invasion (P < .001), and recurrence (P = .013). Subsequent in vitro analyses were conducted using conditioned medium (CM) from immortalized CAFs transfected with siRNA targeting Wnt2. As a result, cancer cell invasion and migration were significantly decreased in the CM from immortalized CAFs transfected with siRNA targeting Wnt2. Our findings indicated that Wnt2 protein released from CAFs enhances CRC cell invasion and migration. In conclusion, Wnt2 secreted by CAFs plays a key role in cancer progression and is a potential therapeutic target for CRC.
Collapse
Affiliation(s)
- Takashi Aizawa
- Department of SurgeryTohoku University Graduate School of MedicineSendaiJapan
| | - Hideaki Karasawa
- Department of SurgeryTohoku University Graduate School of MedicineSendaiJapan
| | - Ryo Funayama
- Department of Cell ProliferationARTTohoku University Graduate School of MedicineSendaiJapan
| | - Matsuyuki Shirota
- Division of Interdisciplinary Medical ScienceARTTohoku University Graduate School of MedicineSendaiJapan
| | - Takashi Suzuki
- Department of Pathology and HistotechnologyTohoku University Graduate School of MedicineSendaiJapan
| | - Shimpei Maeda
- Department of SurgeryTohoku University Graduate School of MedicineSendaiJapan
| | - Hideyuki Suzuki
- Department of SurgeryTohoku University Graduate School of MedicineSendaiJapan
| | - Akihiro Yamamura
- Department of SurgeryTohoku University Graduate School of MedicineSendaiJapan
| | - Takeshi Naitoh
- Department of SurgeryTohoku University Graduate School of MedicineSendaiJapan
| | - Keiko Nakayama
- Department of Cell ProliferationARTTohoku University Graduate School of MedicineSendaiJapan
| | - Michiaki Unno
- Department of SurgeryTohoku University Graduate School of MedicineSendaiJapan
| |
Collapse
|
57
|
Xin X, Yang ST. A Dual Fluorescent 3-D Multicellular Coculture of Breast Cancer MCF-7 and Fibroblast NIH-3T3 Cells for High Throughput Cancer Drug Screening. Biochem Eng J 2019. [DOI: 10.1016/j.bej.2019.05.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
58
|
Abstract
A modified invasion assay using a three-dimensional collagen gel was developed that enables isolation of invasive living cells; it was named the invading cell trapping (iCT) assay. A small cell strainer consisting of a nylon mesh with 40-μm2 pores was used, and collagen gel layers formed across the membrane. Test cells were seeded in the lower gel layer and invasive cells were attracted upward and trapped in the upper gel. After incubation, the collagen gel layers in cell strainers were easily separated and living cells in the gel were counted and analyzed. An advantage of the iCT assay is that it can capture living invasive cells in the upper gel while leaving noninvasive ones in the lower layer. Further enrichment of the two cell populations can be achieved by repeating the assay. Thus, the iCT assay allows comparative analysis of invasive versus noninvasive cells.
Collapse
|
59
|
Yeung TL, Leung CS, Yip KP, Sheng J, Vien L, Bover LC, Birrer MJ, Wong STC, Mok SC. Anticancer Immunotherapy by MFAP5 Blockade Inhibits Fibrosis and Enhances Chemosensitivity in Ovarian and Pancreatic Cancer. Clin Cancer Res 2019; 25:6417-6428. [PMID: 31332047 DOI: 10.1158/1078-0432.ccr-19-0187] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 05/01/2019] [Accepted: 07/09/2019] [Indexed: 01/03/2023]
Abstract
PURPOSE Recent studies demonstrate the role of the tumor microenvironment in tumor progression. However, strategies used to overcome the malignant phenotypes of cancer cells modulated by the microenvironment have not been thoroughly explored. In this study, we evaluated the therapeutic efficacy of a newly developed mAb targeting microfibril-associated protein 5 (MFAP5), which is secreted predominately by cancer-associated fibroblast (CAF), in ovarian and pancreatic cancer models.Experimental Design: MAbs were developed using human MFAP5 recombinant protein as an antigen in mice, and antibodies from hybridoma clones were evaluated for their specificity to human and murine MFAP5. An Octet RED384 system was used to determine the kinetics of binding affinity and the specificity of the antibody clones, which were followed by epitope mapping and functional characterization by in vitro assays. The therapeutic efficacy of a lead anti-MFAP5 antibody clone 130A in tumor suppression was evaluated by ovarian tumor- and pancreatic tumor-bearing mouse models. RESULTS Three hybridoma clones, which produced antibodies with high affinity and specificity to MFAP5, were selected for functional studies. Antibody clone 130A, which recognizes a common epitope shared between human and murine MFAP5 protein, was further selected for in vivo studies. Results showed that clone 130A downregulated MFAP5-induced collagen production in CAFs, suppressed intratumoral microvessel leakiness, and enhanced paclitaxel bioavailability in both ovarian and pancreatic cancer mouse models. CONCLUSIONS These data suggest that MFAP5 blockade using an immunologic approach inhibits fibrosis, induces tumor vessel normalization, and enhances chemosensitivity in ovarian and pancreatic cancer, and can be used as a novel therapeutic agent.
Collapse
Affiliation(s)
- Tsz-Lun Yeung
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Cecilia S Leung
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kay-Pong Yip
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida
| | - Jianting Sheng
- Department of Systems Medicine and Bioengineering, Houston Methodist Cancer Center, Weill Cornell Medical College, Houston, Texas
| | - Long Vien
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Laura C Bover
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas
| | - Michael J Birrer
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama.
| | - Stephen T C Wong
- Department of Systems Medicine and Bioengineering, Houston Methodist Cancer Center, Weill Cornell Medical College, Houston, Texas. .,NCI Center for Modeling Cancer Development, Houston Methodist Research Institute, Houston, Texas
| | - Samuel C Mok
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas. .,The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas
| |
Collapse
|
60
|
Roy R, Morad G, Jedinak A, Moses MA. Metalloproteinases and their roles in human cancer. Anat Rec (Hoboken) 2019; 303:1557-1572. [PMID: 31168956 DOI: 10.1002/ar.24188] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 12/27/2018] [Accepted: 03/04/2019] [Indexed: 02/06/2023]
Abstract
It is now widely appreciated that members of the matrix metalloproteinase (MMP) family of enzymes play a key role in cancer development and progression along with many of the hallmarks associated with them. The activity of these enzymes has been directly implicated in extracellular matrix remodeling, the processing of growth factors and receptors, the modulation of cell migration, proliferation, and invasion, the epithelial to mesenchymal transition, the regulation of immune responses, and the control of angiogenesis. Certain MMP family members have been validated as biomarkers of a variety of human cancers including those of the breast, brain, pancreas, prostate, ovary, and others. The related metalloproteinases, the A disintegrin and metalloproteinases (ADAMs), share a number of these functions as well. Here, we explore these essential metalloproteinases and some of their disease-associated activities in detail as well as some of their complementary translational potential. Anat Rec, 2019. © 2019 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Roopali Roy
- The Vascular Biology Program, Boston Children's Hospital and the Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Golnaz Morad
- The Vascular Biology Program, Boston Children's Hospital and the Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Andrej Jedinak
- The Vascular Biology Program, Boston Children's Hospital and the Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Marsha A Moses
- The Vascular Biology Program, Boston Children's Hospital and the Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
61
|
Amrutkar M, Aasrum M, Verbeke CS, Gladhaug IP. Secretion of fibronectin by human pancreatic stellate cells promotes chemoresistance to gemcitabine in pancreatic cancer cells. BMC Cancer 2019; 19:596. [PMID: 31208372 PMCID: PMC6580453 DOI: 10.1186/s12885-019-5803-1] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 06/06/2019] [Indexed: 02/07/2023] Open
Abstract
Background Gemcitabine remains a cornerstone in chemotherapy of pancreatic ductal adenocarcinoma (PDAC) despite suboptimal clinical effects that are partly due to the development of chemoresistance. Pancreatic stellate cells (PSCs) of the tumor stroma are known to interact with pancreatic cancer cells (PCCs) and influence the progression of PDAC through a complex network of signaling molecules that involve extracellular matrix (ECM) proteins. To understand tumor-stroma interactions regulating chemosensitivity, the role of PSC-secreted fibronectin (FN) in the development of gemcitabine resistance in PDAC was examined. Methods PSC cultures obtained from ten different human PDAC tumors were co-cultured with PCC lines (AsPC-1, BxPC-3, Capan-2, HPAF-II, MIA PaCa-2, PANC-1 and SW-1990) either directly, or indirectly via incubation with PSC-conditioned medium (PSC-CM). Gemcitabine dose response cytotoxicity was determined using MTT based cell viability assays. Protein expression was assessed by western blotting and immunofluorescence. PSC-CM secretome analysis was performed by proteomics-based LC-MS/MS, and FN content in PSC-CM was determined with ELISA. Radiolabeled gemcitabine was used to determine the capacity of PCCs to uptake the drug. Results In both direct and indirect co-culture, PSCs induced varying degrees of resistance to the cytotoxic effects of gemcitabine among all cancer cell lines examined. A variable degree of increased phosphorylation of ERK1/2 was observed across all PCC lines upon incubation with PSC-CM, while activation of AKT was not detected. Secretome analysis of PSC-CM identified 796 different proteins, including several ECM-related proteins such as FN and collagens. Soluble FN content in PSC-CM was detected in the range 175–350 ng/ml. Neither FN nor PSC-CM showed any effect on PCC uptake capacity of gemcitabine. PCCs grown on FN-coated surface displayed higher resistance to gemcitabine compared to cells grown on non-coated surface. Furthermore, a FN inhibitor, synthetic Arg-Gly-Asp-Ser (RGDS) peptide significantly inhibited PSC-CM-induced chemoresistance in PCCs via downregulation of ERK1/2 phosphorylation. Conclusions The findings of this study suggest that FN secreted by PSCs in the ECM plays a key role in the development of resistance to gemcitabine via activation of ERK1/2. FN-blocking agents added to gemcitabine-based chemotherapy might counteract chemoresistance in PDAC and provide better clinical outcomes. Electronic supplementary material The online version of this article (10.1186/s12885-019-5803-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Manoj Amrutkar
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Blindern, 0316, Oslo, Norway. .,Department of Hepato-Pancreato-Biliary Surgery, Institute of Clinical Medicine, University of Oslo, PO Box 1171, Blindern, 0318, Oslo, Norway.
| | - Monica Aasrum
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Blindern, 0316, Oslo, Norway
| | - Caroline S Verbeke
- Department of Pathology, Institute of Clinical Medicine, University of Oslo, Blindern, 0316, Oslo, Norway.,Department of Pathology, Oslo University Hospital Rikshospitalet, Nydalen, 0424, Oslo, Norway
| | - Ivar P Gladhaug
- Department of Hepato-Pancreato-Biliary Surgery, Institute of Clinical Medicine, University of Oslo, PO Box 1171, Blindern, 0318, Oslo, Norway.,Department of Hepato-Pancreato-Biliary Surgery, Oslo University Hospital Rikshospitalet, Nydalen, 0424, Oslo, Norway
| |
Collapse
|
62
|
The Expanding Role of MT1-MMP in Cancer Progression. Pharmaceuticals (Basel) 2019; 12:ph12020077. [PMID: 31137480 PMCID: PMC6630478 DOI: 10.3390/ph12020077] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 05/16/2019] [Accepted: 05/18/2019] [Indexed: 12/21/2022] Open
Abstract
For over 20 years, membrane type 1 matrix metalloproteinase (MT1-MMP) has been recognized as a key component in cancer progression. Initially, the primary roles assigned to MT1-MMP were the activation of proMMP-2 and degradation of fibrillar collagen. Proteomics has revealed a great array of MT1-MMP substrates, and MT1-MMP selective inhibitors have allowed for a more complete mapping of MT1-MMP biological functions. MT1-MMP has extensive sheddase activities, is both a positive and negative regulator of angiogenesis, can act intracellularly and as a transcription factor, and modulates immune responses. We presently examine the multi-faceted role of MT1-MMP in cancer, with a consideration of how the diversity of MT1-MMP behaviors impacts the application of MT1-MMP inhibitors.
Collapse
|
63
|
Han Z, Zhang S, Fujiwara K, Zhang J, Li Y, Liu J, van Zijl PCM, Lu ZR, Zheng L, Liu G. Extradomain-B Fibronectin-Targeted Dextran-Based Chemical Exchange Saturation Transfer Magnetic Resonance Imaging Probe for Detecting Pancreatic Cancer. Bioconjug Chem 2019; 30:1425-1433. [PMID: 30938983 PMCID: PMC6896991 DOI: 10.1021/acs.bioconjchem.9b00161] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
A dextran-peptide conjugate was developed for magnetic resonance (MR) molecular imaging of pancreatic ductal adenocarcinoma (PDAC) through its overexpressed microenvironment biomarker, extradomain-B fibronectin (EDB-FN). This new agent consists of diamagnetic and biocompatible dextran and a targeting peptide. Dextrans can be directly detected by chemical exchange saturation transfer magnetic resonance imaging (CEST MRI) without the need for radionuclide or metallic labeling. In addition, large molecular weight dextran, dextran 10 (MW ∼ 10 kDa), provides an approximately 50 times higher sensitivity per molecule than a single glucose unit. The potential of this highly biocompatible diamagnetic probe is demonstrated in a murine syngeneic allograft PDAC tumor model. The biocompatibility and sensitivity of this new agent clearly show potential for a path to clinical translation.
Collapse
Affiliation(s)
- Zheng Han
- Department of Radiology, Johns Hopkins University, Baltimore, Maryland 21205, United States
- F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland 21205, United States
| | - Shuixing Zhang
- Department of Radiology, Johns Hopkins University, Baltimore, Maryland 21205, United States
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guandong 510630, China
| | - Kenji Fujiwara
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland 21231, United States
| | - Jia Zhang
- Department of Radiology, Johns Hopkins University, Baltimore, Maryland 21205, United States
| | - Yuguo Li
- Department of Radiology, Johns Hopkins University, Baltimore, Maryland 21205, United States
- F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland 21205, United States
| | - Jing Liu
- Department of Radiology, Johns Hopkins University, Baltimore, Maryland 21205, United States
- Radiology Department, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, China
| | - Peter C. M. van Zijl
- Department of Radiology, Johns Hopkins University, Baltimore, Maryland 21205, United States
- F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland 21205, United States
| | - Zheng-Rong Lu
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Lei Zheng
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland 21231, United States
| | - Guanshu Liu
- Department of Radiology, Johns Hopkins University, Baltimore, Maryland 21205, United States
- F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland 21205, United States
| |
Collapse
|
64
|
Wong CW, Han HW, Tien YW, Hsu SH. Biomaterial substrate-derived compact cellular spheroids mimicking the behavior of pancreatic cancer and microenvironment. Biomaterials 2019; 213:119202. [PMID: 31132644 DOI: 10.1016/j.biomaterials.2019.05.013] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 05/05/2019] [Accepted: 05/08/2019] [Indexed: 12/19/2022]
Abstract
Pancreatic stromal cells especially pancreatic stellate cells (PSCs) play a critical role in the progression of human pancreatic ductal adenocarcinoma (PDAC). However, the exact interaction between cancer cells and PSCs remains to be elucidated in order to develop more effective therapeutic approaches to treat PDAC. The microenvironment of PDAC shows higher hyaluronan (HA) levels, which is associated with poor prognosis of PDAC patients. In the current study, an efficient three-dimensional tumor spheroid model for PDAC was established. The pancreatic cancer cells and PSCs were co-cultured on hyaluronan grafted chitosan (CS-HA) coated plates to generate 3D tumor-like co-spheroids. The pancreatic cancer cells and PSCs (1:9 ratio) co-cultured on CS-HA coated plates were assembled into tumor-like co-spheroids with 3D core-shell structure in 48 h. These spheroids displayed potent in vitro tumorigenicity such as up-regulated expression of stemness and migration markers. The migration rate of cancer cells in spheroids (from 1:9 cell ratio) was much faster (3.2-fold) than that of cancer cells alone. Meanwhile, this unique co-spheroidal cancer cell structure with the outer wrap of PSCs contributed to the chemo-resistance of pancreatic cancer cells to gemcitabine as well as sensitivity to the combined gemcitabine and Abraxane treatment in vitro. The metastatic nature of the spheroids was confirmed by the zebrafish xenograft model in vivo. The compact and dynamic pancreatic cancer-PSC co-spheroids generated by the unique 3D co-culture platform on CS-HA biomaterials can mimic the PSC-constituting microenvironment of PDAC and demonstrate the chemo-resistant, invasive, and metastatic phenotypes. They have potential applications in personalized and high-throughput drug screening.
Collapse
Affiliation(s)
- Chui-Wei Wong
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei, Taiwan
| | - Hao-Wei Han
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei, Taiwan
| | - Yu-Wen Tien
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan.
| | - Shan-Hui Hsu
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei, Taiwan; Research and Development Center for Medical Devices, National Taiwan University, Taipei, Taiwan; Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan.
| |
Collapse
|
65
|
Chen YT, Chen FW, Chang TH, Wang TW, Hsu TP, Chi JY, Hsiao YW, Li CF, Wang JM. Hepatoma-derived growth factor supports the antiapoptosis and profibrosis of pancreatic stellate cells. Cancer Lett 2019; 457:180-190. [PMID: 31078734 DOI: 10.1016/j.canlet.2019.05.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 03/30/2019] [Accepted: 05/02/2019] [Indexed: 12/13/2022]
Abstract
Pancreatic cancer is refractory and is characterized by extensively surrounding and intratumor fibrotic reactions that are contributed by activated pancreatic stellate cells (PSCs). Herein, we show that CCAAT/enhancer-binding protein δ (CEBPD) responds to transforming growth factor-β1 (TGF-β1) through reciprocal loop regulation and that activated hypoxia inducible factor-1α (HIF-1α) further contributes to the upregulation of the hepatoma-derived growth factor (HDGF) gene. Secreted HDGF contributes to the antiapoptosis of PSCs and consequently leads to the synthesis and deposition of extracellular matrix proteins for stabilizing PSC/pancreatic cancer cell (PCC) tumor foci. This result agrees with the observation that severe stromal growth positively correlated with stromal HDGF and CEBPD expression in pancreatic cancer specimens. Collectively, the identification of the TGF-β1-activated CEBPD/HIF-1α/HDGF axis provides new insights into novel discoveries of HDGF in the antiapoptosis and profibrosis of PSCs and the outgrowth of PCCs.
Collapse
Affiliation(s)
- Yi-Ting Chen
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan; Medical Research Department, Chi Mei Medical Center, Tainan, Taiwan
| | - Feng-Wei Chen
- Institute of Basic Medical Science, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Tsung-Hao Chang
- Institute of Basic Medical Science, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Tso-Wen Wang
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Teng-Po Hsu
- Department of Life Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Jhih-Ying Chi
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Wei Hsiao
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Chien-Feng Li
- Medical Research Department, Chi Mei Medical Center, Tainan, Taiwan; Department of Pathology, Chi Mei Medical Center, Tainan, Taiwan.
| | - Ju-Ming Wang
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan; International Research Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan, Taiwan; Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
66
|
Huet E, Jaroz C, Nguyen HQ, Belkacemi Y, de la Taille A, Stavrinides V, Whitaker H. Stroma in normal and cancer wound healing. FEBS J 2019; 286:2909-2920. [PMID: 30958920 DOI: 10.1111/febs.14842] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 02/18/2019] [Accepted: 04/05/2019] [Indexed: 12/28/2022]
Abstract
It is currently believed that stroma, the connective framework of biological tissues, plays a central role in normal wound healing and in cancer. In both these contexts, stromal cellular components such as activated fibroblasts interact with complex protein networks that include growth factors, structural protein or proteinases in order to initiate and sustain an extensive remodelling process. However, although this process is usually spatially and temporally self-limited, it is unregulated in the case of cancer and leads to uncontrolled cell proliferation and invasion within tissues, metastasis and therapeutic resistance. In this review, we outline the role of stroma in normal healing, cancer and post radiotherapy, with a particular focus on the crosstalk between normal or cancer cells and fibroblasts. Understanding these mechanisms is particularly important as several stromal components have been proposed as potential therapeutic targets.
Collapse
Affiliation(s)
- Eric Huet
- Université Paris-Est, UPEC, Créteil, France.,INSERM, U955, Equipe 7, Créteil, France.,Molecular Diagnostics and Therapeutics Group, Charles Bell House, Division of Surgery and Interventional Science, University College London, UK
| | | | | | - Yazid Belkacemi
- INSERM, U955, Equipe 7, Créteil, France.,AP-HP, Hôpitaux Universitaires Henri Mondor, Service d'oncologie-radiothérapie et Centre Sein Henri Mondor Créteil, France
| | - Alexandre de la Taille
- INSERM, U955, Equipe 7, Créteil, France.,AP-HP, Hôpitaux Universitaires Henri Mondor, Service d'urologie, Créteil, France
| | - Vasilis Stavrinides
- Molecular Diagnostics and Therapeutics Group, Charles Bell House, Division of Surgery and Interventional Science, University College London, UK
| | - Hayley Whitaker
- Molecular Diagnostics and Therapeutics Group, Charles Bell House, Division of Surgery and Interventional Science, University College London, UK
| |
Collapse
|
67
|
Schnittert J, Bansal R, Mardhian DF, van Baarlen J, Östman A, Prakash J. Integrin α11 in pancreatic stellate cells regulates tumor stroma interaction in pancreatic cancer. FASEB J 2019; 33:6609-6621. [PMID: 30808244 DOI: 10.1096/fj.201802336r] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the deadliest tumor due to its highly abundant tumor stroma. Pancreatic stellate cells (PSCs) are considered precursor cells of cancer-associated fibroblasts (CAFs), which induce tumor progression, invasion, and metastasis. In this study, we investigated the role of integrin subunit α (ITGA) 11, the receptor for collagen type I, in tumor stroma interaction. Clinical sample analysis showed that ITGA11 was overexpressed by CAFs in PDAC stroma, as shown with colocalization immunostaining with α-smooth muscle actin. In contrast, there was no expression in healthy pancreas. Public transcriptomic data confirmed a reduced expression of ITGA11 in healthy pancreas and adjacent nontumoral tissues compared with human tumor tissues. Primary human PSCs (hPSCs) activated with either TGF-β or pancreatic cancer cell (PANC-1)-conditioned medium (CM) resulted in the significant up-regulation of ITGA11 and various CAF markers. Furthermore, short hairpin RNA (shRNA)-mediated stable ITGA11 knockdown (shITGA11) in hPSCs significantly inhibited TGF-β- and PANC-1 CM-mediated activation at both gene and protein levels of extracellular matrix, cytokines, and adhesion molecules. Additionally, shITGA11 hPSCs had a reduced migration and contractility compared with shRNA control (shCTR) PSCs. Furthermore, we investigated the effect of ITGA11 on the paracrine effects of hPSCs. Interestingly, the CM from shITGA11 hPSCs, activated with either TGF-β or PANC-1 CM, caused tumor cells to migrate and invade lesser compared with their counterpart, activated shCTR PSCs. In summary, this study presents ITGA11 as an interesting stromal therapeutic target that plays a crucial role in the regulation of the differentiation of PSCs into CAFs and paracrine effects.-Schnittert, J., Bansal, R., Mardhian, D. F., van Baarlen, J., Östman, A., Prakash, J. Integrin α11 in pancreatic stellate cells regulates tumor stroma interaction in pancreatic cancer.
Collapse
Affiliation(s)
- Jonas Schnittert
- Department of Biomaterials, Science, and Technology, TechMed Centre, Targeted Therapeutics Section, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| | - Ruchi Bansal
- Department of Biomaterials, Science, and Technology, TechMed Centre, Targeted Therapeutics Section, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| | - Deby F Mardhian
- Department of Biomaterials, Science, and Technology, TechMed Centre, Targeted Therapeutics Section, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| | - Joop van Baarlen
- Laboratory Pathology East Netherlands (LabPON), Hengelo, The Netherlands
| | - Arne Östman
- Department of Oncology-Pathology, Cancer Centre Karolinska, Karolinska Institutet, Stockholm, Sweden
| | - Jai Prakash
- Department of Biomaterials, Science, and Technology, TechMed Centre, Targeted Therapeutics Section, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands.,Department of Oncology-Pathology, Cancer Centre Karolinska, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
68
|
Durymanov M, Kroll C, Permyakova A, Reineke J. Role of Endocytosis in Nanoparticle Penetration of 3D Pancreatic Cancer Spheroids. Mol Pharm 2019; 16:1074-1082. [DOI: 10.1021/acs.molpharmaceut.8b01078] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Mikhail Durymanov
- Department of Pharmaceutical Sciences, College of Pharmacy, South Dakota State University, Brookings, South Dakota 57007, United States
| | - Christian Kroll
- Department of Pharmaceutical Sciences, College of Pharmacy, South Dakota State University, Brookings, South Dakota 57007, United States
| | - Anastasia Permyakova
- Department of Pharmaceutical Sciences, College of Pharmacy, South Dakota State University, Brookings, South Dakota 57007, United States
| | - Joshua Reineke
- Department of Pharmaceutical Sciences, College of Pharmacy, South Dakota State University, Brookings, South Dakota 57007, United States
| |
Collapse
|
69
|
Thomas D, Radhakrishnan P. Tumor-stromal crosstalk in pancreatic cancer and tissue fibrosis. Mol Cancer 2019; 18:14. [PMID: 30665410 PMCID: PMC6341551 DOI: 10.1186/s12943-018-0927-5] [Citation(s) in RCA: 277] [Impact Index Per Article: 46.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 12/20/2018] [Indexed: 12/12/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a devastating disease with high morbidity and mortality worldwide. To date, limited therapeutic achievements targeting cell proliferation and related mechanisms has led researchers to focus on the microenvironment where pancreatic cancers develop. The anomalous proliferation of stromal cells, such as pancreatic stellate cells, and an increased deposition of altered matrix proteins create an environment that facilitates tumor growth, metastasis and drug resistance. Here, we summarize our understanding of recent advances in research about the role of fibrosis in pancreatic cancer progression, with particular emphasize on the involvement of fibrotic machineries such as wound healing, extra cellular matrix degradation, and epithelial-to-mesenchymal transition. The precise influence of these mechanisms on the biological behaviors and growth of cancer cells has great impact on clinical therapy and therefore deserves more attention. We also discuss the role of various stromal components in conferring drug resistance to PDAC which further worsening the pessimistic disease prognosis. A more in depth understanding of cancer-stroma crosstalk within the tumor microenvironment and stroma based clinical and translational therapies may provide new therapeutic strategies for the prevention of pancreatic cancer progression.
Collapse
Affiliation(s)
- Divya Thomas
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, 986805 Nebraska Medical Center, Omaha, NE, 68198-6805, USA
| | - Prakash Radhakrishnan
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, 986805 Nebraska Medical Center, Omaha, NE, 68198-6805, USA.
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA.
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
70
|
Yu Q, Qiu Y, Chen X, Wang X, Mei L, Wu H, Liu K, Liu Y, Li M, Zhang Z, He Q. Chemotherapy priming of the Pancreatic Tumor Microenvironment Promotes Delivery and Anti-Metastasis Efficacy of Intravenous Low-Molecular-Weight Heparin-Coated Lipid-siRNA Complex. Am J Cancer Res 2019; 9:355-368. [PMID: 30809279 PMCID: PMC6376180 DOI: 10.7150/thno.29137] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 11/19/2018] [Indexed: 02/07/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a type of malignant tumor with high lethality. Its high tumor cell-density and large variety of extracellular matrix (ECM) components present major barriers for drug delivery. Methods: Paclitaxel-loaded PEGylated liposomes (PTX-Lip) were used as a tumor-priming agent to induce tumor cell apoptosis and decrease the abundance of ECM to promote cellular uptake and tumor delivery of nanodrugs. Paclitaxel exerts anti-cancer effects but, paradoxically, exacerbates cancer metastasis and drug resistance by increasing the expression of apoptotic B-cell lymphoma-2 protein (BCL-2). Thus, low-molecular-weight heparin-coated lipid-siRNA complex (LH-Lip/siBCL-2) was constructed to inhibit cancer metastasis and silence BCL-2 by BCL-2 siRNA (siBCL-2). Results: Significant tumor growth inhibition efficacy was observed, accompanied by obvious inhibition of cancer metastasis in vivo. Conclusion: These results suggested our sequential delivery of PTX-Lip and LH-Lip/siBCL-2 might provide a practical approach for PDAC or other ECM-rich tumors.
Collapse
|
71
|
Liu J, Tian L, Zhang R, Dong Z, Wang H, Liu Z. Collagenase-Encapsulated pH-Responsive Nanoscale Coordination Polymers for Tumor Microenvironment Modulation and Enhanced Photodynamic Nanomedicine. ACS APPLIED MATERIALS & INTERFACES 2018; 10:43493-43502. [PMID: 30468076 DOI: 10.1021/acsami.8b17684] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
The abundant tumor extracellular matrix (ECM) could result in insufficient tumor retention and ineffective intratumor penetration of therapeutic agents as well as an acidic and hypoxic tumor microenvironment (TME), leading to unsatisfactory therapeutic outcomes for many types of therapies. Therefore, developing strategies to modulate the TME by selectively degrading the condensed ECM may be helpful to improve existing cancer therapies. Herein, collagenase (CLG)-encapsulated nanoscale coordination polymers (NCPs) are synthesized based on Mn2+ and an acid-sensitive benzoic-imine organic linker and then modified by polyethylene glycol (PEG). Upon intravenous (iv) injection, these CLG@NCP-PEG nanoparticles show efficient accumulation within the tumor, in which CLG would be released because of the collapse of NCP structures within the acidic TME. The released CLG enzyme could then specifically degrade collagens, the major component of ECM, leading to a loosened ECM structure, enhanced tumor perfusion, and relieved hypoxia. As a result, the second wave of nanoparticles, chlorin e6 (Ce6)-loaded liposomes (liposome@Ce6), would exhibit enhanced retention and penetration within the tumor. Such phenomena together with relieved tumor hypoxia could then lead to greatly enhanced photodynamic therapeutic effect of liposome@Ce6 for mice pretreated with CLG@NCP-PEG. Our work thus presents a unique strategy for TME modulation using pH-responsive NCPs as smart enzyme carriers.
Collapse
Affiliation(s)
- Jingjing Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices , Soochow University , Suzhou , Jiangsu 215123 , China
| | - Longlong Tian
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices , Soochow University , Suzhou , Jiangsu 215123 , China
| | - Rui Zhang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices , Soochow University , Suzhou , Jiangsu 215123 , China
| | - Ziliang Dong
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices , Soochow University , Suzhou , Jiangsu 215123 , China
| | - Hairong Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices , Soochow University , Suzhou , Jiangsu 215123 , China
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices , Soochow University , Suzhou , Jiangsu 215123 , China
| |
Collapse
|
72
|
Quantitative Method to Track Proteolytic Invasion in 3D Collagen. Methods Mol Biol 2018. [PMID: 30378053 DOI: 10.1007/978-1-4939-8879-2_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Since many tumors are associated with a pronounced collagen-rich stromal reaction, there is increasing interest in understanding mechanisms by which cancer cells invade through the collagen barrier. Here we describe a quantitative method to track cell invasion in 3D collagen I gels. We analyze invasion by quantifying proteolytic tracks generated by invading cancer cells through a 3D collagen microenvironment. We provide a detailed protocol for this quantitative assay, which can be used to characterize signaling pathways that regulate invasion in the 3D microenvironment.
Collapse
|
73
|
Durymanov M, Kroll C, Permyakova A, O'Neill E, Sulaiman R, Person M, Reineke J. Subcutaneous Inoculation of 3D Pancreatic Cancer Spheroids Results in Development of Reproducible Stroma-Rich Tumors. Transl Oncol 2018; 12:180-189. [PMID: 30554606 PMCID: PMC6295361 DOI: 10.1016/j.tranon.2018.10.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 10/02/2018] [Accepted: 10/02/2018] [Indexed: 12/21/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a deadly disease characterized by high expression of extracellular matrix in tumor tissue, which contributes to chemoresistance and poor prognosis. Here, we developed 3D pancreatic cancer spheroids, based on pancreatic cancer cells and fibroblast co-culture, which demonstrate innate desmoplastic properties and stay poorly permeable for model nanoparticles. Our study revealed that establishment of tumors by transplantation of spheroids significantly improved subcutaneous xenograft model of PDAC, which stays the most widely used animal model for testing of new drugs and drug delivery approaches. Spheroid based tumors abundantly produced different extracellular matrix (ECM) components including collagen I, fibronectin, laminin and hyaluronic acid. These tumors were highly reproducible with excellent uniformity in terms of ECM architecture recapitulating clinical PDAC tumors, whereas in more common cell based xenografts a significant intertumor heterogeneity in extracellular matrix production was found. Moreover, spheroid based xenografts demonstrated higher expression of pro-fibrotic and pro-survival PDAC hallmarks in opposite to cell based counterparts. We believe that future development of this model will provide an effective instrument for testing of anti-cancer drugs with improved predictive value.
Collapse
Affiliation(s)
- Mikhail Durymanov
- Department of Pharmaceutical Sciences, South Dakota State University, Brookings, SD
| | - Christian Kroll
- Department of Pharmaceutical Sciences, South Dakota State University, Brookings, SD
| | - Anastasia Permyakova
- Department of Pharmaceutical Sciences, South Dakota State University, Brookings, SD
| | | | - Raed Sulaiman
- Department of Pathology and Laboratory Medicine, Avera McKennan Hospital, Sioux Falls, SD
| | | | - Joshua Reineke
- Department of Pharmaceutical Sciences, South Dakota State University, Brookings, SD.
| |
Collapse
|
74
|
Ahn B, Chae YS, Kim CH, Lee Y, Lee JH, Kim JY. Tumor microenvironmental factors have prognostic significances in advanced gastric cancer. APMIS 2018; 126:814-821. [PMID: 30264431 DOI: 10.1111/apm.12889] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 08/20/2018] [Indexed: 11/28/2022]
Abstract
Tumor microenvironment is important in the progression and survival of cancer cells. We evaluated the prognostic significance of tumor stroma percentage (TSP), Klintrup-Mäkinen (KM) grade, which reflects the density of inflammatory cells of the tumor, and Glasgow microenvironment score (GMS), a combination of TSP and KM grade, in advanced gastric cancers. A total of 196 pT3 and pT4 gastric cancers were histologically evaluated using TSP, KM grade, and GMS. These were correlated with other clinicopathologic factors including patients' survival. High TSP (78 cases), low KM grade (124 cases), and higher GMS (score 0, 72 cases; 1, 53 cases; and 2, 71 cases) were correlated with poor differentiation, diffuse type, presence of lymphovascular invasion, perineural invasion, and lymph node metastasis. High TSP was significantly correlated with low KM grade (p < 0.001). High TSP (HR, 3.079, 95% CI, 1.612-5.883, p = 0.001), low KM grade (3.201, 1.774-5.776, p < 0.001), and higher GMS (12.274, 3.684-40.895, p < 0.001) were independent poor prognostic factors. TSP, KM grade, and GMS are significantly associated with clinicopathologic behavior and patients' survival. Assessing these factors is a feasible and cost-effective way to identify tumor microenvironment with different biological features and prognosis of gastric cancer patients.
Collapse
Affiliation(s)
- Bokyung Ahn
- Department of Pathology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Korea
| | - Yang-Seok Chae
- Department of Pathology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Korea
| | - Chul Hwan Kim
- Department of Pathology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Korea
| | - Youngseok Lee
- Department of Pathology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Korea
| | - Jeong Hyeon Lee
- Department of Pathology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Korea
| | - Joo Young Kim
- Department of Pathology, Nowon Eulji Medical Center, Eulji University, Seoul, Korea
| |
Collapse
|
75
|
Mardhian DF, Storm G, Bansal R, Prakash J. Nano-targeted relaxin impairs fibrosis and tumor growth in pancreatic cancer and improves the efficacy of gemcitabine in vivo. J Control Release 2018; 290:1-10. [PMID: 30287265 DOI: 10.1016/j.jconrel.2018.09.031] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 09/11/2018] [Accepted: 09/30/2018] [Indexed: 12/18/2022]
Abstract
Cancer-associated fibroblasts (CAFs), are the key effector cells in pancreatic ductal adenocarcinoma (PDAC), known to induce tumor growth and progression. Pancreatic stellate cells (PSCs) are the precursors of CAFs in PDAC that secrete abundant extracellular matrix, growth factors and cytokines. In this study, we targeted human relaxin-2 (RLX), an endogenous hormone, to PSCs to inhibit their differentiation into CAF-like myofibroblasts. RLX significantly inhibited TGF-β induced PSCs differentiation by inhibiting pSmad2 signaling pathway. In vitro in primary human PSCs (hPSCs), treatment with RLX dose-dependently inhibited the migration, contraction, and protein expression of alpha smooth muscle actin and collagen I These data demonstrate that RLX can regulate hPSCs activation in vitro. However, RLX has several drawbacks i.e. poor pharmacokinetics and systemic vasodilation, that limits its preclinical and clinical application. Thus, we designed and successfully synthesized a nanoparticle system by chemically conjugating RLX to superparamagnetic iron oxide nanoparticle (SPION) to improve its pharmacokinetics. Interestingly, we found RLX-SPION to be more efficacious compared to free RLX in vitro. Significantly, we observed RLX-SPION retarded the tumor growth by itself and also potentiated the effect of gemcitabine in a subcutaneous co-injection (Panc1 and hPSCs) tumor model. The treatment resulted in significant inhibition in tumor growth, which was attributed to reduced collagen I (ECM), desmin (hPSC marker) and CD31 (endothelial marker) expression. In contrast, free RLX showed no significant effects. Altogether, this study presents a novel therapeutic approach against tumor stroma using RLX-SPION to achieve an effective treatment against pancreatic tumor.
Collapse
Affiliation(s)
- Deby F Mardhian
- Section - Targeted Therapeutics, Department of Biomaterials Science and Technology, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| | - Gert Storm
- Section - Targeted Therapeutics, Department of Biomaterials Science and Technology, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands; Department of Pharmaceutics, Utrecht University, Utrecht, The Netherlands
| | - Ruchi Bansal
- Section - Targeted Therapeutics, Department of Biomaterials Science and Technology, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| | - Jai Prakash
- Section - Targeted Therapeutics, Department of Biomaterials Science and Technology, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands; ScarTec Therapeutics BV, Enschede, The Netherlands.
| |
Collapse
|
76
|
Ebine K, Kumar K, Pham TN, Shields MA, Collier KA, Shang M, DeCant BT, Urrutia R, Hwang RF, Grimaldo S, Principe DR, Grippo PJ, Bentrem DJ, Munshi HG. Interplay between interferon regulatory factor 1 and BRD4 in the regulation of PD-L1 in pancreatic stellate cells. Sci Rep 2018; 8:13225. [PMID: 30185888 PMCID: PMC6125340 DOI: 10.1038/s41598-018-31658-1] [Citation(s) in RCA: 28] [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: 05/01/2018] [Accepted: 08/21/2018] [Indexed: 01/18/2023] Open
Abstract
The fibrotic reaction is a characteristic feature of human pancreatic ductal adenocarcinoma (PDAC) tumors. It is associated with activation and proliferation of pancreatic stellate cells (PSCs), which are key regulators of fibrosis in vivo. While there is increasing interest in the regulation of PD-L1 expression in cancer and immune cells, the expression and regulation of PD-L1 in other stromal cells, such as PSCs, has not been fully evaluated. Here we show that PSCs in vitro express higher PD-L1 mRNA and protein levels compared to the levels present in PDAC cells. We show that inhibitors targeting bromodomain and extra-terminal (BET) proteins and BRD4 knockdown decrease interferon-γ (IFN-γ)-induced PD-L1 expression in PSCs. We also show that c-MYC, one of the well-established targets of BET inhibitors, does not mediate IFN-γ-regulated PD-L1 expression in PSCs. Instead we show that interferon regulatory factor 1 (IRF1) mediates IFN-γ-induced PD-L1 expression in PSCs. Finally, while we show that BET inhibitors do not regulate IFN-γ-induced IRF1 expression in PSCs, BET inhibitors decrease binding of IRF1 and BRD4 to the PD-L1 promoter. Together, these results demonstrate the interplay between IRF1 and BRD4 in the regulation of PD-L1 in PSCs.
Collapse
Affiliation(s)
- Kazumi Ebine
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Jesse Brown VA Medical Center, Chicago, IL, USA
| | - Krishan Kumar
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
- The Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA.
| | - Thao N Pham
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Mario A Shields
- Cold Spring Harbor Laboratory, Cold Spring Harbor, Cold Spring, NY, USA
| | - Katharine A Collier
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Meng Shang
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Jesse Brown VA Medical Center, Chicago, IL, USA
| | - Brian T DeCant
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Raul Urrutia
- Laboratory of Epigenetics and Chromatin Dynamics, Division of Gastroenterology and Hepatology, Department of Internal Medicine, Epigenomics Translational Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA
| | - Rosa F Hwang
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sam Grimaldo
- Department of Medicine, University of Illinois, Chicago, IL, USA
| | | | - Paul J Grippo
- Department of Medicine, University of Illinois, Chicago, IL, USA
| | - David J Bentrem
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Jesse Brown VA Medical Center, Chicago, IL, USA
- The Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA
| | - Hidayatullah G Munshi
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
- Jesse Brown VA Medical Center, Chicago, IL, USA.
- The Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA.
| |
Collapse
|
77
|
Podolsky MJ, Gupta D, Ha A, Ta R, Khalifeh-Soltani A, McKleroy W, Datta R, Sheppard D, Atabai K. Cell division cycle 7 kinase is a negative regulator of cell-mediated collagen degradation. Am J Physiol Lung Cell Mol Physiol 2018; 315:L360-L370. [PMID: 29792348 DOI: 10.1152/ajplung.00144.2018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Although extensive work has delineated many of the mechanisms of extracellular matrix (ECM) production, far less is known about pathways that regulate ECM degradation. This is particularly true of cellular internalization and degradation of matrix, which play an underappreciated role in ECM metabolism and lung fibrosis. For example, genetic perturbation of this pathway leads to exacerbated fibrosis in experimental animal models. In this work, we present the results of an unbiased screen of Drosophila phagocytes that yielded multiple genes that, when silenced, led to increased collagen uptake. We further describe the function of cell division cycle 7 kinase (CDC7) as a specific suppressor of collagen uptake. We show that the genetic or pharmacological inhibition of CDC7 results in increased expression of the collagen endocytic receptor Endo180. Chromobox 5 (CBX5) is a putative target of CDC7, and genetic silencing of CBX5 also results in increased Endo180 and collagen uptake. Finally, CRISPR-mediated activation of Endo180 expression results in increased collagen uptake, suggesting that CDC7 regulates collagen internalization through increased Endo180 expression. Targeting the regulatory elements of the collagen degradative machinery may be a useful therapeutic approach in diseases of fibrosis or malignancy.
Collapse
Affiliation(s)
- Michael J Podolsky
- Department of Medicine, Lung Biology Center, Cardiovascular Research Institute, University of California, San Francisco, California
| | - Deepti Gupta
- Department of Medicine, Lung Biology Center, Cardiovascular Research Institute, University of California, San Francisco, California
| | - Arnold Ha
- Department of Medicine, Lung Biology Center, Cardiovascular Research Institute, University of California, San Francisco, California
| | - Ryan Ta
- Department of Medicine, Lung Biology Center, Cardiovascular Research Institute, University of California, San Francisco, California
| | - Amin Khalifeh-Soltani
- Department of Medicine, Lung Biology Center, Cardiovascular Research Institute, University of California, San Francisco, California
| | - William McKleroy
- Department of Medicine, Lung Biology Center, Cardiovascular Research Institute, University of California, San Francisco, California
| | - Ritwik Datta
- Department of Medicine, Lung Biology Center, Cardiovascular Research Institute, University of California, San Francisco, California
| | - Dean Sheppard
- Department of Medicine, Lung Biology Center, Cardiovascular Research Institute, University of California, San Francisco, California
| | - Kamran Atabai
- Department of Medicine, Lung Biology Center, Cardiovascular Research Institute, University of California, San Francisco, California
| |
Collapse
|
78
|
Felician FF, Xia C, Qi W, Xu H. Collagen from Marine Biological Sources and Medical Applications. Chem Biodivers 2018. [DOI: 10.1002/cbdv.201700557] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Fatuma Felix Felician
- The Engineering Research Center of Peptide Drug Discovery and Development; China Pharmaceutical University; Nanjing 211198 Jiangsu Province P. R. China
| | - Chunlei Xia
- The Engineering Research Center of Peptide Drug Discovery and Development; China Pharmaceutical University; Nanjing 211198 Jiangsu Province P. R. China
| | - Weiyan Qi
- The Engineering Research Center of Peptide Drug Discovery and Development; China Pharmaceutical University; Nanjing 211198 Jiangsu Province P. R. China
- Department of Marine Pharmacy; College of Life Science and Technology; P. R. China Pharmaceutical University; Nanjing 211198 Jiangsu Province P. R. China
| | - Hanmei Xu
- The Engineering Research Center of Peptide Drug Discovery and Development; China Pharmaceutical University; Nanjing 211198 Jiangsu Province P. R. China
- Department of Marine Pharmacy; College of Life Science and Technology; P. R. China Pharmaceutical University; Nanjing 211198 Jiangsu Province P. R. China
| |
Collapse
|
79
|
Biondani G, Zeeberg K, Greco MR, Cannone S, Dando I, Dalla Pozza E, Mastrodonato M, Forciniti S, Casavola V, Palmieri M, Reshkin SJ, Cardone RA. Extracellular matrix composition modulates PDAC parenchymal and stem cell plasticity and behavior through the secretome. FEBS J 2018; 285:2104-2124. [PMID: 29660229 DOI: 10.1111/febs.14471] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 03/05/2018] [Accepted: 04/06/2018] [Indexed: 12/17/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancers. Its aggressiveness is driven by an intense fibrotic desmoplastic reaction in which the increasingly collagen I-rich extracellular matrix (ECM) and several cell types, including cancer stem cells (CSCs), create a tumor-supportive environment. However, how ECM composition regulates CSC dynamics and their relationship with the principle parenchymal tumor population to promote early invasive growth is not yet characterized. For this, we utilized a platform of 3D organotypic cultures composed of laminin-rich Matrigel, representative of an early tumor, plus increasing concentrations of collagen I to simulate malignant stroma progression. As ECM collagen I increases, CSCs progress from a rapidly growing, vascular phenotype to a slower growing, avascular phase, while maintaining their endothelial-like gene signatures. This transition is supported autocrinically by the CSCs and paracrinically by the parenchymal cells via their ECM-dependent secretomes. Indeed, when growing on an early tumor ECM, the CSCs are dedicated toward the preparation of a vascular niche by (a) activating their growth program, (b) secreting high levels of proangiogenic factors which stimulate both angiogenesis and vasculogenic mimicry, and (c) overexpressing VEGFR-2, which is activated by VEGF secreted by both the CSC and parenchymal cells. On Matrigel, the more differentiated parenchymal tumor cell population had reduced growth but a high invasive capacity. This concerted high local invasion of parenchymal cells into the CSC-derived vascular network suggests that a symbiotic relationship between the parenchymal cells and the CSCs underlies the initiation and maintenance of early PDAC infiltration and metastasis.
Collapse
Affiliation(s)
- Giulia Biondani
- Department of Neuroscience, Biomedicine and Movement, Biochemistry Section, University of Verona, Italy
| | - Katrine Zeeberg
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, Italy
| | - Maria Raffaella Greco
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, Italy
| | - Stefania Cannone
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, Italy
| | - Ilaria Dando
- Department of Neuroscience, Biomedicine and Movement, Biochemistry Section, University of Verona, Italy
| | - Elisa Dalla Pozza
- Department of Neuroscience, Biomedicine and Movement, Biochemistry Section, University of Verona, Italy
| | | | - Stefania Forciniti
- Department of Neuroscience, Biomedicine and Movement, Biochemistry Section, University of Verona, Italy
| | - Valeria Casavola
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, Italy
| | - Marta Palmieri
- Department of Neuroscience, Biomedicine and Movement, Biochemistry Section, University of Verona, Italy
| | - Stephan Joel Reshkin
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, Italy
| | - Rosa Angela Cardone
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, Italy
| |
Collapse
|
80
|
Liu HY, Korc M, Lin CC. Biomimetic and enzyme-responsive dynamic hydrogels for studying cell-matrix interactions in pancreatic ductal adenocarcinoma. Biomaterials 2018; 160:24-36. [PMID: 29353105 PMCID: PMC5815383 DOI: 10.1016/j.biomaterials.2018.01.012] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 01/03/2018] [Accepted: 01/06/2018] [Indexed: 01/18/2023]
Abstract
The tumor microenvironment (TME) governs all aspects of cancer progression and in vitro 3D cell culture platforms are increasingly developed to emulate the interactions between components of the stromal tissues and cancer cells. However, conventional cell culture platforms are inadequate in recapitulating the TME, which has complex compositions and dynamically changing matrix mechanics. In this study, we developed a dynamic gelatin-hyaluronic acid hybrid hydrogel system through integrating modular thiol-norbornene photopolymerization and enzyme-triggered on-demand matrix stiffening. In particular, gelatin was dually modified with norbornene and 4-hydroxyphenylacetic acid to render this bioactive protein photo-crosslinkable (through thiol-norbornene gelation) and responsive to tyrosinase-triggered on-demand stiffening (through HPA dimerization). In addition to the modified gelatin that provides basic cell adhesive motifs and protease cleavable sequences, hyaluronic acid (HA), an essential tumor matrix, was modularly and covalently incorporated into the cell-laden gel network. We systematically characterized macromer modification, gel crosslinking, as well as enzyme-triggered stiffening and degradation. We also evaluated the influence of matrix composition and dynamic stiffening on pancreatic ductal adenocarcinoma (PDAC) cell fate in 3D. We found that either HA-containing matrix or a dynamically stiffened microenvironment inhibited PDAC cell growth. Interestingly, these two factors synergistically induced cell phenotypic changes that resembled cell migration and/or invasion in 3D. Additional mRNA expression array analyses revealed changes unique to the presence of HA, to a stiffened microenvironment, or to the combination of both. Finally, we presented immunostaining and mRNA expression data to demonstrate that these irregular PDAC cell phenotypes were a result of matrix-induced epithelial-mesenchymal transition (EMT).
Collapse
Affiliation(s)
- Hung-Yi Liu
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Murray Korc
- Department of Medicine and Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Indiana University Melvin and Bren Simon Cancer Center, and The Pancreatic Cancer Signature Center, Indianapolis, IN 46202, USA
| | - Chien-Chi Lin
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA; Indiana University Melvin and Bren Simon Cancer Center, and The Pancreatic Cancer Signature Center, Indianapolis, IN 46202, USA; Department of Biomedical Engineering, Purdue School of Engineering & Technology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA.
| |
Collapse
|
81
|
Schnittert J, Heinrich MA, Kuninty PR, Storm G, Prakash J. Reprogramming tumor stroma using an endogenous lipid lipoxin A4 to treat pancreatic cancer. Cancer Lett 2018; 420:247-258. [DOI: 10.1016/j.canlet.2018.01.072] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Revised: 12/25/2017] [Accepted: 01/22/2018] [Indexed: 12/18/2022]
|
82
|
Hamada S, Masamune A. Elucidating the link between collagen and pancreatic cancer: what's next? Expert Rev Gastroenterol Hepatol 2018; 12:315-317. [PMID: 29495889 DOI: 10.1080/17474124.2018.1448268] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- Shin Hamada
- a Division of Gastroenterology , Tohoku University Graduate School of Medicine , Sendai , Japan
| | - Atsushi Masamune
- a Division of Gastroenterology , Tohoku University Graduate School of Medicine , Sendai , Japan
| |
Collapse
|
83
|
Jayatilaka H, Giri A, Karl M, Aifuwa I, Trenton NJ, Phillip JM, Khatau S, Wirtz D. EB1 and cytoplasmic dynein mediate protrusion dynamics for efficient 3-dimensional cell migration. FASEB J 2018; 32:1207-1221. [PMID: 29097501 PMCID: PMC5893312 DOI: 10.1096/fj.201700444rr] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 10/23/2017] [Indexed: 11/12/2022]
Abstract
Microtubules have long been implicated to play an integral role in metastatic disease, for which a critical step is the local invasion of tumor cells into the 3-dimensional (3D) collagen-rich stromal matrix. Here we show that cell migration of human cancer cells uses the dynamic formation of highly branched protrusions that are composed of a microtubule core surrounded by cortical actin, a cytoskeletal organization that is absent in cells on 2-dimensional (2D) substrates. Microtubule plus-end tracking protein End-binding 1 and motor protein dynein subunits light intermediate chain 2 and heavy chain 1, which do not regulate 2D migration, critically modulate 3D migration by affecting RhoA and thus regulate protrusion branching through differential assembly dynamics of microtubules. An important consequence of this observation is that the commonly used cancer drug paclitaxel is 100-fold more effective at blocking migration in a 3D matrix than on a 2D matrix. This work reveals the central role that microtubule dynamics plays in powering cell migration in a more pathologically relevant setting and suggests further testing of therapeutics targeting microtubules to mitigate migration.-Jayatilaka, H., Giri, A., Karl, M., Aifuwa, I., Trenton, N. J., Phillip, J. M., Khatau, S., Wirtz, D. EB1 and cytoplasmic dynein mediate protrusion dynamics for efficient 3-dimensional cell migration.
Collapse
Affiliation(s)
- Hasini Jayatilaka
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland, USA
- Johns Hopkins Physical Sciences–Oncology Center, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Anjil Giri
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland, USA
- Johns Hopkins Physical Sciences–Oncology Center, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Michelle Karl
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland, USA
- Johns Hopkins Physical Sciences–Oncology Center, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Ivie Aifuwa
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland, USA
- Johns Hopkins Physical Sciences–Oncology Center, The Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Jude M. Phillip
- Division of Hematology and Oncology, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Shyam Khatau
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Denis Wirtz
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, Maryland, USA
- Johns Hopkins Physical Sciences–Oncology Center, The Johns Hopkins University, Baltimore, Maryland, USA
- Department of Pathology, The Johns Hopkins School of Medicine, Baltimore, Maryland, USA
- Department of Oncology, The Johns Hopkins School of Medicine, Baltimore, Maryland, USA
- Sydney Kimmel Comprehensive Cancer Center, The Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
84
|
Lee JH, Kim SK, Khawar IA, Jeong SY, Chung S, Kuh HJ. Microfluidic co-culture of pancreatic tumor spheroids with stellate cells as a novel 3D model for investigation of stroma-mediated cell motility and drug resistance. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:4. [PMID: 29329547 PMCID: PMC5767067 DOI: 10.1186/s13046-017-0654-6] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 11/27/2017] [Indexed: 12/24/2022]
Abstract
BACKGROUND Pancreatic stellate cells (PSCs), a major component of the tumor microenvironment in pancreatic cancer, play roles in cancer progression as well as drug resistance. Culturing various cells in microfluidic (microchannel) devices has proven to be a useful in studying cellular interactions and drug sensitivity. Here we present a microchannel plate-based co-culture model that integrates tumor spheroids with PSCs in a three-dimensional (3D) collagen matrix to mimic the tumor microenvironment in vivo by recapitulating epithelial-mesenchymal transition and chemoresistance. METHODS A 7-channel microchannel plate was prepared using poly-dimethylsiloxane (PDMS) via soft lithography. PANC-1, a human pancreatic cancer cell line, and PSCs, each within a designated channel of the microchannel plate, were cultured embedded in type I collagen. Expression of EMT-related markers and factors was analyzed using immunofluorescent staining or Proteome analysis. Changes in viability following exposure to gemcitabine and paclitaxel were measured using Live/Dead assay. RESULTS PANC-1 cells formed 3D tumor spheroids within 5 days and the number of spheroids increased when co-cultured with PSCs. Culture conditions were optimized for PANC-1 cells and PSCs, and their appropriate interaction was confirmed by reciprocal activation shown as increased cell motility. PSCs under co-culture showed an increased expression of α-SMA. Expression of EMT-related markers, such as vimentin and TGF-β, was higher in co-cultured PANC-1 spheroids compared to that in mono-cultured spheroids; as was the expression of many other EMT-related factors including TIMP1 and IL-8. Following gemcitabine exposure, no significant changes in survival were observed. When paclitaxel was combined with gemcitabine, a growth inhibitory advantage was prominent in tumor spheroids, which was accompanied by significant cytotoxicity in PSCs. CONCLUSIONS We demonstrated that cancer cells grown as tumor spheroids in a 3D collagen matrix and PSCs co-cultured in sub-millimeter proximity participate in mutual interactions that induce EMT and drug resistance in a microchannel plate. Microfluidic co-culture of pancreatic tumor spheroids with PSCs may serve as a useful model for studying EMT and drug resistance in a clinically relevant manner.
Collapse
Affiliation(s)
- Ji-Hyun Lee
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seul-Ki Kim
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Iftikhar Ali Khawar
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Su-Yeong Jeong
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seok Chung
- School of Mechanical Engineering, College of Engineering, Korea University, Seoul, Republic of Korea
| | - Hyo-Jeong Kuh
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea. .,Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-ku, Seoul, 06591, Republic of Korea.
| |
Collapse
|
85
|
Couvineau A, Dayot S, Nicole P, Gratio V, Rebours V, Couvelard A, Voisin T. The Anti-tumoral Properties of Orexin/Hypocretin Hypothalamic Neuropeptides: An Unexpected Therapeutic Role. Front Endocrinol (Lausanne) 2018; 9:573. [PMID: 30319552 PMCID: PMC6170602 DOI: 10.3389/fendo.2018.00573] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 09/10/2018] [Indexed: 02/05/2023] Open
Abstract
Orexins (OxA and OxB) also termed hypocretins are hypothalamic neuropeptides involved in central nervous system (CNS) to control the sleep/wake process which is mediated by two G protein-coupled receptor subtypes, OX1R, and OX2R. Beside these central effects, orexins also play a role in various peripheral organs such as the intestine, pancreas, adrenal glands, kidney, adipose tissue and reproductive tract.In the past few years, an unexpected anti-tumoral role of orexins mediated by a new signaling pathway involving the presence of two immunoreceptor tyrosine-based inhibitory motifs (ITIM) in both orexin receptors subtypes, the recruitment of the phosphotyrosine phosphatase SHP2 and the induction of mitochondrial apoptosis has been elucidated. In the present review, we will discuss the anti-tumoral effect of orexin/OXR system in colon, pancreas, prostate and other cancers, and its interest as a possible therapeutic target.
Collapse
|
86
|
Abstract
The extracellular matrix (ECM) is a meshwork of proteins and carbohydrates that supports many biological structures and processes, from tissue development and elasticity to preserve the structures of entire organs. In each organ, the composition of the ECM is distinct. It is a remarkably active three-dimensional structure that is continuously undergoing remodeling to regulate tissue homeostasis. This review aims to explain the role of ECM proteins in the remodeling process in different types of disease. The hardening of the ECM (desmoplasia), as well as its manipulation, induction, and impairment in regulation of its composition can play a role in several diseases, examples of which are chronic obstructive pulmonary disease, pancreatic ductal adenocarcinoma, spinal cord injury, progression and metastasis of breast cancer, and neurodegenerative condition in the brain such as Alzheimer's disease. Remodeling is also associated with diet-induced insulin resistance in many metabolic tissues. A greater comprehension of the way in which the ECM regulates organ structure and function and of how ECM remodeling affects the development of diseases may lead to the improvement and discovery of new treatments.
Collapse
Affiliation(s)
- Hala Salim Sonbol
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21332, Saudi Arabia
| |
Collapse
|
87
|
Principe DR, DeCant B, Diaz AM, Mangan RJ, Hwang R, Lowy A, Shetuni BB, Sreekumar BK, Chung C, Bentrem DJ, Munshi HG, Jung B, Grippo PJ, Bishehsari F. PEDF inhibits pancreatic tumorigenesis by attenuating the fibro-inflammatory reaction. Oncotarget 2017; 7:28218-34. [PMID: 27058416 PMCID: PMC5053722 DOI: 10.18632/oncotarget.8587] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 03/17/2016] [Indexed: 12/19/2022] Open
Abstract
Pancreatic cancer is characterized by a pronounced fibro-inflammatory reaction that has been shown to contribute to cancer progression. Previous reports have demonstrated that pigment epithelium-derived factor (PEDF) has potent tumor suppressive effects in pancreatic cancer, though little is known about the mechanisms by which PEDF limits pancreatic tumorigenesis. We therefore employed human specimens, as well as mouse and in vitro models, to explore the effects of PEDF upon the pancreatic microenvironment. We found that PEDF expression is decreased in human pancreatic cancer samples compared to non-malignant tissue. Furthermore, PEDF-deficient patients displayed increased intratumoral inflammation/fibrosis. In mice, genetic ablation of PEDF increased cerulein-induced inflammation and fibrosis, and similarly enhanced these events in the background of oncogenic KRAS. In vitro, recombinant PEDF neutralized macrophage migration as well as inhibited macrophage-induced proliferation of tumor cells. Additionally, recombinant PEDF suppressed the synthesis of pro-inflammatory/pro-fibrotic cytokines both in vivo and in vitro, and reduced collagen I deposition and TGFβ synthesis by pancreatic stellate cells, consistent with reduced fibrosis. Combined, our results demonstrate that PEDF limits pancreatic cancer progression by attenuating the fibro-inflammatory reaction, and makes restoration of PEDF signaling a potential therapeutic approach to study in pancreatic cancer.
Collapse
Affiliation(s)
| | - Brian DeCant
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Andrew M Diaz
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Riley J Mangan
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Rosa Hwang
- Department of Surgical Oncology, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Andrew Lowy
- Department of Surgery, University of California San Diego, San Diego, CA, USA
| | | | - Bharath K Sreekumar
- Department of Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Chuhan Chung
- Department of Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - David J Bentrem
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Hidayatullah G Munshi
- Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Barbara Jung
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Paul J Grippo
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Faraz Bishehsari
- Department of Medicine, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
88
|
Liang C, Shi S, Meng Q, Liang D, Ji S, Zhang B, Qin Y, Xu J, Ni Q, Yu X. Complex roles of the stroma in the intrinsic resistance to gemcitabine in pancreatic cancer: where we are and where we are going. Exp Mol Med 2017; 49:e406. [PMID: 29611542 PMCID: PMC5750480 DOI: 10.1038/emm.2017.255] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 07/23/2017] [Accepted: 08/07/2017] [Indexed: 01/18/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is among the most devastating human malignancies. The poor clinical outcome in PDAC is partly attributed to a growth-permissive tumor microenvironment. In the PDAC microenvironment, the stroma is characterized by the development of extensive fibrosis, with stromal components outnumbering pancreatic cancer cells. Each of the components within the stroma has a distinct role in conferring chemoresistance to PDAC, and intrinsic chemoresistance has further worsened this pessimistic prognosis. The nucleoside analog gemcitabine (GEM) is usually the recommended first-line chemotherapeutic agent for PDAC patients and is given alone or in combination with other agents. The mechanisms of intrinsic resistance to GEM are an active area of ongoing research. This review highlights the important role the complex structure of stroma in PDAC plays in the intrinsic resistance to GEM and discusses whether antistroma therapy improves the efficacy of GEM. The addition of antistroma therapy combined with GEM is expected to be a novel therapeutic strategy with significant survival benefits for PDAC patients.
Collapse
Affiliation(s)
- Chen Liang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Si Shi
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Qingcai Meng
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Dingkong Liang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Shunrong Ji
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Bo Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Yi Qin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Jin Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Quanxing Ni
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| |
Collapse
|
89
|
Puls TJ, Tan X, Whittington CF, Voytik-Harbin SL. 3D collagen fibrillar microstructure guides pancreatic cancer cell phenotype and serves as a critical design parameter for phenotypic models of EMT. PLoS One 2017; 12:e0188870. [PMID: 29190794 PMCID: PMC5708668 DOI: 10.1371/journal.pone.0188870] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 11/14/2017] [Indexed: 12/14/2022] Open
Abstract
Pancreatic cancer, one of the deadliest cancers, is characterized by high rates of metastasis and intense desmoplasia, both of which are associated with changes in fibrillar type I collagen composition and microstructure. Epithelial to mesenchymal transition (EMT), a critical step of metastasis, also involves a change in extracellular matrix (ECM) context as cells detach from basement membrane (BM) and engage interstitial matrix (IM). The objective of this work was to develop and apply an in-vitro three-dimensional (3D) tumor-ECM model to define how ECM composition and biophysical properties modulate pancreatic cancer EMT. Three established pancreatic ductal adenocarcinoma (PDAC) lines were embedded within 3D matrices prepared with type I collagen Oligomer (IM) at various fibril densities to control matrix stiffness or Oligomer and Matrigel combined at various ratios while maintaining constant matrix stiffness. Evaluation of cell morphology and protein expression at both the cellular- and population-levels revealed a spectrum of matrix-driven EMT phenotypes that were dependent on ECM composition and architecture as well as initial PDAC phenotype. In general, exposure to fibrillar IM was sufficient to drive EMT, with cells displaying spindle-shaped morphology and mesenchymal markers, and non-fibrillar BM promoted more epithelial behavior. When cultured within low density Oligomer, only a subpopulation of epithelial BxPC-3 cells displayed EMT while mesenchymal MiaPaCa-2 cells displayed more uniform spindle-shaped morphologies and mesenchymal marker expression. Interestingly, as IM fibril density increased, associated changes in spatial constraints and matrix stiffness resulted in all PDAC lines growing as tight clusters; however mesenchymal marker expression was maintained. Collectively, the comparison of these results to other in-vitro tumor models highlights the role of IM fibril microstructure in guiding EMT heterogeneity and showcases the potential of standardized 3D matrices such as Oligomer to serve as robust platforms for mechanistic study of metastasis and creation of predictive drug screening models.
Collapse
Affiliation(s)
- T. J. Puls
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, United States of America
| | - Xiaohong Tan
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, United States of America
| | - Catherine F. Whittington
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, United States of America
- Department of Oncology, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Sherry L. Voytik-Harbin
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, United States of America
- Department of Basic Medical Sciences, Purdue University, West Lafayette, Indiana, United States of America
- * E-mail:
| |
Collapse
|
90
|
Kim JH, Shin BC, Park WS, Lee J, Kuh HJ. Antifibrotic effects of pentoxifylline improve the efficacy of gemcitabine in human pancreatic tumor xenografts. Cancer Sci 2017; 108:2470-2477. [PMID: 28940685 PMCID: PMC5715266 DOI: 10.1111/cas.13405] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Revised: 09/12/2017] [Accepted: 09/13/2017] [Indexed: 02/06/2023] Open
Abstract
We investigated the combinatorial effects of pentoxifylline (PTX) on the efficacy of gemcitabine (GEM) in a human pancreatic tumor xenograft model. PTX significantly improved the efficacy of GEM, as shown by a 50% reduction in tumor growth rate at 4 weeks of treatment compared with that in animals given GEM alone. The fluorescent drug doxorubicin (DOX) was used to test whether drug delivery was improved by PTX, contributing to the improved efficacy of GEM. PTX given for 2 weeks prior to giving DOX improved drug distribution by 1.8‐ to 2.2‐fold with no changes in vessel density, suggesting that improvement in drug delivery was not related to the vascular mechanism. Instead, collagen I content in tumor stroma was significantly reduced, as was the expression of alpha‐smooth muscle actin of cancer‐associated fibroblasts and connective tissue growth factor (CTGF) by PTX pretreatment. Overall, our data demonstrated that increased efficacy of GEM by PTX was associated with improved drug delivery to tumor tissue, which may be attributed to decreased expression of CTGF and subsequent reduction in the stromal collagen matrix in the pancreatic ductal adenocarcinoma tumor. These results support the usefulness of PTX in combination with chemotherapy for targeting drug delivery barriers associated with the stromal matrix, which should be further evaluated for clinical development.
Collapse
Affiliation(s)
- Jung Ho Kim
- Department of Biomedicine & Health Science, The Catholic University of Korea, Seoul, Korea
| | - Byung Cheol Shin
- Bio/Drug Discovery Division, Korea Research Institute of Chemical Technology, Daejeon, Korea
| | - Won Sang Park
- Department of Pathology, The Catholic University of Korea, Seoul, Korea
| | - Jaehwi Lee
- College of Pharmacy, Chung-Ang University, Seoul, Korea
| | - Hyo-Jeong Kuh
- Department of Biomedicine & Health Science, The Catholic University of Korea, Seoul, Korea.,Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
91
|
Tzanakakis G, Kavasi RM, Voudouri K, Berdiaki A, Spyridaki I, Tsatsakis A, Nikitovic D. Role of the extracellular matrix in cancer-associated epithelial to mesenchymal transition phenomenon. Dev Dyn 2017; 247:368-381. [PMID: 28758355 DOI: 10.1002/dvdy.24557] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 06/20/2017] [Accepted: 07/08/2017] [Indexed: 12/14/2022] Open
Abstract
The epithelial to mesenchymal transition (EMT) program is a crucial component in the processes of morphogenesis and embryonic development. The transition of epithelial to mesenchymal phenotype is associated with numerous structural and functional changes, including loss of cell polarity and tight cell-cell junctions, the acquisition of invasive abilities, and the expression of mesenchymal proteins. The switch between the two phenotypes is involved in human pathology and is crucial for cancer progression. Extracellular matrices (ECMs) are multi-component networks that surround cells in tissues. These networks are obligatory for cell survival, growth, and differentiation as well as tissue organization. Indeed, the ECM suprastructure, in addition to its supportive role, can process and deliver a plethora of signals to cells, which ultimately regulate their behavior. Importantly, the ECM derived signals are critically involved in the process of EMT during tumorigenesis. This review discusses the multilayer interaction between the ECM and the EMT process, focusing on contributions of discrete mediators, a strategy that may identify novel potential target molecules. Developmental Dynamics 247:368-381, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- George Tzanakakis
- Laboratory of Anatomy-Histology-Embryology, School of Medicine, University of Crete, Heraklion, Greece
| | - Rafaela-Maria Kavasi
- Laboratory of Anatomy-Histology-Embryology, School of Medicine, University of Crete, Heraklion, Greece
| | - Kallirroi Voudouri
- Laboratory of Anatomy-Histology-Embryology, School of Medicine, University of Crete, Heraklion, Greece
| | - Aikaterini Berdiaki
- Laboratory of Anatomy-Histology-Embryology, School of Medicine, University of Crete, Heraklion, Greece
| | - Ioanna Spyridaki
- Laboratory of Anatomy-Histology-Embryology, School of Medicine, University of Crete, Heraklion, Greece
| | - Aristidis Tsatsakis
- Laboratory of Toxicology, School of Medicine, University of Crete, Heraklion, Greece
| | - Dragana Nikitovic
- Laboratory of Anatomy-Histology-Embryology, School of Medicine, University of Crete, Heraklion, Greece
| |
Collapse
|
92
|
The Role of Tumor Microenvironment in Chemoresistance: To Survive, Keep Your Enemies Closer. Int J Mol Sci 2017; 18:ijms18071586. [PMID: 28754000 PMCID: PMC5536073 DOI: 10.3390/ijms18071586] [Citation(s) in RCA: 299] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 07/16/2017] [Accepted: 07/19/2017] [Indexed: 12/12/2022] Open
Abstract
Chemoresistance is a leading cause of morbidity and mortality in cancer and it continues to be a challenge in cancer treatment. Chemoresistance is influenced by genetic and epigenetic alterations which affect drug uptake, metabolism and export of drugs at the cellular levels. While most research has focused on tumor cell autonomous mechanisms of chemoresistance, the tumor microenvironment has emerged as a key player in the development of chemoresistance and in malignant progression, thereby influencing the development of novel therapies in clinical oncology. It is not surprising that the study of the tumor microenvironment is now considered to be as important as the study of tumor cells. Recent advances in technological and analytical methods, especially ‘omics’ technologies, has made it possible to identify specific targets in tumor cells and within the tumor microenvironment to eradicate cancer. Tumors need constant support from previously ‘unsupportive’ microenvironments. Novel therapeutic strategies that inhibit such microenvironmental support to tumor cells would reduce chemoresistance and tumor relapse. Such strategies can target stromal cells, proteins released by stromal cells and non-cellular components such as the extracellular matrix (ECM) within the tumor microenvironment. Novel in vitro tumor biology models that recapitulate the in vivo tumor microenvironment such as multicellular tumor spheroids, biomimetic scaffolds and tumor organoids are being developed and are increasing our understanding of cancer cell-microenvironment interactions. This review offers an analysis of recent developments on the role of the tumor microenvironment in the development of chemoresistance and the strategies to overcome microenvironment-mediated chemoresistance. We propose a systematic analysis of the relationship between tumor cells and their respective tumor microenvironments and our data show that, to survive, cancer cells interact closely with tumor microenvironment components such as mesenchymal stem cells and the extracellular matrix.
Collapse
|
93
|
Collagen-derived proline promotes pancreatic ductal adenocarcinoma cell survival under nutrient limited conditions. Nat Commun 2017; 8:16031. [PMID: 28685754 PMCID: PMC5504351 DOI: 10.1038/ncomms16031] [Citation(s) in RCA: 299] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 05/23/2017] [Indexed: 12/12/2022] Open
Abstract
Tissue architecture contributes to pancreatic ductal adenocarcinoma (PDAC) phenotypes. Cancer cells within PDAC form gland-like structures embedded in a collagen-rich meshwork where nutrients and oxygen are scarce. Altered metabolism is needed for tumour cells to survive in this environment, but the metabolic modifications that allow PDAC cells to endure these conditions are incompletely understood. Here we demonstrate that collagen serves as a proline reservoir for PDAC cells to use as a nutrient source when other fuels are limited. We show PDAC cells are able to take up collagen fragments, which can promote PDAC cell survival under nutrient limited conditions, and that collagen-derived proline contributes to PDAC cell metabolism. Finally, we show that proline oxidase (PRODH1) is required for PDAC cell proliferation in vitro and in vivo. Collectively, our results indicate that PDAC extracellular matrix represents a nutrient reservoir for tumour cells highlighting the metabolic flexibility of this cancer. Cancer cells adapt their metabolism to survive limited nutrient availability. Here, the authors show that in conditions of limited glucose or glutamine availability, pancreatic ductal adenocarcinoma cells can use collagen-derived proline to foster the TCA cycle and allow cell survival both in vitro and in vivo.
Collapse
|
94
|
Abstract
Fibrosis is defined as excess deposition of extracellular matrix, resulting in tissue scarring and organ dysfunction. It is estimated that 45% of deaths in the developed world are due to fibrosis-induced organ failure. Despite the well-accepted role of fibrosis in the pathogenesis of numerous diseases, there are only two US Food and Drug Administration–approved anti-fibrotic therapies, both of which are currently restricted to the treatment of pulmonary fibrosis. Thus, organ fibrosis represents a massive unmet medical need. Here, we review recent findings suggesting that an epigenetic regulatory protein, BRD4, is a nodal effector of organ fibrosis, and we highlight the potential of small-molecule BRD4 inhibitors for the treatment of diverse fibrotic diseases.
Collapse
Affiliation(s)
- Matthew S Stratton
- Department of Medicine, Division of Cardiology and Consortium for Fibrosis Research & Translation, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Saptarsi M Haldar
- Gladstone Institutes and Department of Medicine, Division of Cardiology, University of California San Francisco School of Medicine, San Francisco, CA, USA
| | - Timothy A McKinsey
- Department of Medicine, Division of Cardiology and Consortium for Fibrosis Research & Translation, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
95
|
Pulmonary delivery of nanoparticle chemotherapy for the treatment of lung cancers: challenges and opportunities. Acta Pharmacol Sin 2017; 38:782-797. [PMID: 28504252 DOI: 10.1038/aps.2017.34] [Citation(s) in RCA: 182] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Accepted: 02/04/2017] [Indexed: 12/11/2022]
Abstract
Lung cancer is the second most prevalent and the deadliest among all cancer types. Chemotherapy is recommended for lung cancers to control tumor growth and to prolong patient survival. Systemic chemotherapy typically has very limited efficacy as well as severe systemic adverse effects, which are often attributed to the distribution of anticancer drugs to non-targeted sites. In contrast, inhalation routes permit the delivery of drugs directly to the lungs providing high local concentrations that may enhance the anti-tumor effect while alleviating systemic adverse effects. Preliminary studies in animals and humans have suggested that most inhaled chemotherapies are tolerable with manageable pulmonary adverse effects, including cough and bronchospasm. Promoting the deposition of anticancer drugs in tumorous cells and minimizing access to healthy lung cells can further augment the efficacy and reduce the risk of local toxicities caused by inhaled chemotherapy. Sustained release and tumor localization characteristics make nanoparticle formulations a promising candidate for the inhaled delivery of chemotherapeutic agents against lung cancers. However, the physiology of respiratory tracts and lung clearance mechanisms present key barriers for the effective deposition and retention of inhaled nanoparticle formulations in the lungs. Recent research has focused on the development of novel formulations to maximize lung deposition and to minimize pulmonary clearance of inhaled nanoparticles. This article systematically reviews the challenges and opportunities for the pulmonary delivery of nanoparticle formulations for the treatment of lung cancers.
Collapse
|
96
|
Zhou ZH, Ji CD, Xiao HL, Zhao HB, Cui YH, Bian XW. Reorganized Collagen in the Tumor Microenvironment of Gastric Cancer and Its Association with Prognosis. J Cancer 2017. [PMID: 28638462 PMCID: PMC5479253 DOI: 10.7150/jca.18466] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Collagen components in the tumor microenvironment substantially influence cancer pathogenesis and progression. Nevertheless, in gastric cancer, collagen status and its prognostic role remain unclear. Using picrosirius red staining and immunohistochemistry, we found that collagen deposition was significantly increased in gastric cancer when compared with non-neoplastic tissues, and in cancer stroma, more immature collagen components were present, suggesting a qualitative change. Furthermore, the morphology of collagen fibers could be weakly, moderately or strongly changed in gastric cancer; when weakly or moderately changed, they appeared similar to normal collagen fibers, except for a higher linearization and density; when strongly changed, they were thicker and less eosinophilic, sharply differently from their normal counterparts. In addition, we found abundant myofibroblasts and elevated expression of lysyl oxidase-like 2 (the enzyme that mediates crosslinking of collagen molecules) in cancer stroma, which might contribute to the increased collagen deposition and crosslinking. Last, five collagen architectural parameters (alignment, density, width, length and straightness) were analyzed with second harmonic generation imaging, a highly specific technology for detection of collagen fibers, and our data indicated that all the parameters were significantly increased in the tumor microenvironment. Of the five parameters, collagen width was the most powerful parameter in predicting 5-year overall survival, and increased collagen width was associated with reduced survival. The prognostic value of collagen width was superior to traditional clinicopathological parameters, and this was validated in two unrelated gastric cancer cohorts that contained 225 and 151 patients. Collectively, the collagen status (content, maturity, morphology and architecture) was profoundly reorganized in the tumor microenvironment of gastric cancer, and collagen width could serve as a valuable prognostic indicator.
Collapse
Affiliation(s)
- Zhi-Hua Zhou
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Cheng-Dong Ji
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Hua-Liang Xiao
- Department of Pathology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Hai-Bin Zhao
- Department of Pathology, The 101 Hospital of PLA, Wuxi, Jiangsu Province, China
| | - You-Hong Cui
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Xiu-Wu Bian
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China.,Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| |
Collapse
|
97
|
Zhou ZH, Ji CD, Zhu J, Xiao HL, Zhao HB, Cui YH, Bian XW. The prognostic value and pathobiological significance of Glasgow microenvironment score in gastric cancer. J Cancer Res Clin Oncol 2017; 143:883-894. [PMID: 28180998 DOI: 10.1007/s00432-017-2346-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 01/17/2017] [Indexed: 12/22/2022]
Abstract
PURPOSE To evaluate the prognostic value and pathobiological significance of Glasgow microenvironment score (GMS), a parameter based on tumor stroma percentage and inflammatory cell infiltration, in gastric cancer. METHODS A total of 225 cases of gastric cancer were histologically reviewed, and GMS was evaluated for each case. The association between GMS and patients' survival was investigated. Then the relationship between GMS and mismatch repair (MMR) status, Epstein-Barr virus (EBV) infection were determined using immunohistochemistry (IHC) and in situ hybridization, and the expression of PD1/PD-L1 was examined. Furthermore, the amount of cancer-associated fibroblasts (CAFs), the content and maturity of collagen components were detected using IHC, Picrosirius Red staining and second harmonic generation imaging. RESULTS GMS was significantly associated with clinical outcomes of gastric cancer, and multivariate analysis indicated that GMS was an independent factor (HR 1.725, P = 0.002). Low GMS was a manifestation of better prognosis and inflammatory tumor microenvironment, which was related to MMR deficiency (P = 0.042) and EBV infection (P = 0.032), and within this microenvironment, expression of PD-L1 in carcinoma cells (P = 0.030) or in inflammatory cells (P = 0.029) was significantly higher. In contrast, high GMS linked to a poorer survival and desmoplastic stroma, in which there existed markedly increased CAFs and collagen deposition. CONCLUSION GMS can serve as a useful prognostic factor for gastric cancer, and according to GMS, the tumor microenvironment in this cancer type may be partially classified as inflammatory or desmoplastic microenvironment that possesses different pathobiological features.
Collapse
Affiliation(s)
- Zhi-Hua Zhou
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Gaotanyan 30, Chongqing, 400038, China
- Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
- Department of Pathology, The 101 Hospital of People's Liberation Army, Wuxi, Jiangsu Province, China
| | - Cheng-Dong Ji
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Gaotanyan 30, Chongqing, 400038, China
- Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Jiang Zhu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Gaotanyan 30, Chongqing, 400038, China
| | - Hua-Liang Xiao
- Department of Pathology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Hai-Bin Zhao
- Department of Pathology, The 101 Hospital of People's Liberation Army, Wuxi, Jiangsu Province, China
| | - You-Hong Cui
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Gaotanyan 30, Chongqing, 400038, China
- Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Xiu-Wu Bian
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Gaotanyan 30, Chongqing, 400038, China.
- Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China.
| |
Collapse
|
98
|
Narayanan S, Loganathan G, Dhanasekaran M, Tucker W, Patel A, Subhashree V, Mokshagundam S, Hughes MG, Williams SK, Balamurugan AN. Intra-islet endothelial cell and β-cell crosstalk: Implication for islet cell transplantation. World J Transplant 2017; 7:117-128. [PMID: 28507914 PMCID: PMC5409911 DOI: 10.5500/wjt.v7.i2.117] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 02/28/2017] [Accepted: 03/24/2017] [Indexed: 02/05/2023] Open
Abstract
The intra-islet microvasculature is a critical interface between the blood and islet endocrine cells governing a number of cellular and pathophysiological processes associated with the pancreatic tissue. A growing body of evidence indicates a strong functional and physical interdependency of β-cells with endothelial cells (ECs), the building blocks of islet microvasculature. Intra-islet ECs, actively regulate vascular permeability and appear to play a role in fine-tuning blood glucose sensing and regulation. These cells also tend to behave as “guardians”, controlling the expression and movement of a number of important immune mediators, thereby strongly contributing to the physiology of islets. This review will focus on the molecular signalling and crosstalk between the intra-islet ECs and β-cells and how their relationship can be a potential target for intervention strategies in islet pathology and islet transplantation.
Collapse
|
99
|
Knapinska AM, Estrada CA, Fields GB. The Roles of Matrix Metalloproteinases in Pancreatic Cancer. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 148:339-354. [PMID: 28662827 DOI: 10.1016/bs.pmbts.2017.03.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Matrix metalloproteinases (MMPs) have long been implicated for roles in cancer initiation, tumor growth, and metastasis. However, pancreatic cancer clinical trials using broad-based MMP inhibitors were discouraging. To better evaluate the use of MMP inhibitors in pancreatic cancer, (a) more precise roles of individual MMPs in pancreatic cancer needed to be determined and (b) animal models that more accurately represented human pancreatic cancer needed to be developed. The last decade has seen substantial progress in both areas. MT1-MMP has been recognized as a critical mediator of several steps in pancreatic cancer progression, while MMP-9 appears to be an antitarget when considering pancreatic cancer therapies.
Collapse
Affiliation(s)
| | | | - Gregg B Fields
- Florida Atlantic University, Jupiter, FL, United States; The Scripps Research Institute/Scripps Florida, Jupiter, FL, United States.
| |
Collapse
|
100
|
Turunen SP, Tatti-Bugaeva O, Lehti K. Membrane-type matrix metalloproteases as diverse effectors of cancer progression. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:1974-1988. [PMID: 28390905 DOI: 10.1016/j.bbamcr.2017.04.002] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 04/02/2017] [Accepted: 04/03/2017] [Indexed: 12/12/2022]
Abstract
Membrane-type matrix metalloproteases (MT-MMP) are pivotal regulators of cell invasion, growth and survival. Tethered to the cell membranes by a transmembrane domain or GPI-anchor, the six MT-MMPs can exert these functions via cell surface-associated extracellular matrix degradation or proteolytic protein processing, including shedding or release of signaling receptors, adhesion molecules, growth factors and other pericellular proteins. By interactions with signaling scaffold or cytoskeleton, the C-terminal cytoplasmic tail of the transmembrane MT-MMPs further extends their functionality to signaling or structural relay. MT-MMPs are differentially expressed in cancer. The most extensively studied MMP14/MT1-MMP is induced in various cancers along malignant transformation via pathways activated by mutations in tumor suppressors or proto-oncogenes and changes in tumor microenvironment including cellular heterogeneity, extracellular matrix composition, tissue oxygenation, and inflammation. Classically such induction involves transcriptional programs related to epithelial-to-mesenchymal transition. Besides inhibition by endogenous tissue inhibitors, MT-MMP activities are spatially and timely regulated at multiple levels by microtubular vesicular trafficking, dimerization/oligomerization, other interactions and localization in the actin-based invadosomes, in both tumor and the stroma. The functions of MT-MMPs are multifaceted within reciprocal cellular responses in the evolving tumor microenvironment, which poses the importance of these proteases beyond the central function as matrix scissors, and necessitates us to rethink MT-MMPs as dynamic signaling proteases of cancer. This article is part of a Special Issue entitled: Matrix Metalloproteinases edited by Rafael Fridman.
Collapse
Affiliation(s)
- S Pauliina Turunen
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Nobels väg 16, SE-17177 Stockholm, Sweden
| | - Olga Tatti-Bugaeva
- Research Programs Unit, Genome-Scale Biology and Haartman Institute, University of Helsinki, and Helsinki University Hospital, P.O. Box 63, FI-00014 Helsinki, Finland
| | - Kaisa Lehti
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Nobels väg 16, SE-17177 Stockholm, Sweden; Research Programs Unit, Genome-Scale Biology and Haartman Institute, University of Helsinki, and Helsinki University Hospital, P.O. Box 63, FI-00014 Helsinki, Finland; K. Albin Johansson Foundation, Finnish Cancer Institute, P.O. Box 63, FI-00014, Helsinki, Finland.
| |
Collapse
|