1
|
Bhatia R, Chang J, Munoz JL, Walker ND. Forging New Therapeutic Targets: Efforts of Tumor Derived Exosomes to Prepare the Pre-Metastatic Niche for Cancer Cell Dissemination and Dormancy. Biomedicines 2023; 11:1614. [PMID: 37371709 PMCID: PMC10295689 DOI: 10.3390/biomedicines11061614] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/23/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
Tumor-derived exosomes play a multifaceted role in preparing the pre-metastatic niche, promoting cancer dissemination, and regulating cancer cell dormancy. A brief review of three types of cells implicated in metastasis and an overview of other types of extracellular vesicles related to metastasis are described. A central focus of this review is on how exosomes influence cancer progression throughout metastatic disease. Exosomes are crucial mediators of intercellular communication by transferring their cargo to recipient cells, modulating their behavior, and promoting tumor pro-gression. First, their functional role in cancer cell dissemination in the peripheral blood by facilitating the establishment of a pro-angiogenic and pro-inflammatory niche is described during organotro-pism and in lymphatic-mediated metastasis. Second, tumor-derived exosomes can transfer molecular signals that induce cell cycle arrest, dormancy, and survival pathways in disseminated cells, promoting a dormant state are reviewed. Third, several studies highlight exosome involvement in maintaining cellular dormancy in the bone marrow endosteum. Finally, the clinical implications of exosomes as biomarkers or diagnostic tools for cancer progression are also outlined. Understanding the complex interplay between tumor-derived exosomes and the pre-metastatic niche is crucial for developing novel therapeutic strategies to target metastasis and prevent cancer recurrence. To that end, several examples of how exosomes or other nanocarriers are used as a drug delivery system to inhibit cancer metastasis are discussed. Strategies are discussed to alter exosome cargo content for better loading capacity or direct cell targeting by integrins. Further, pre-clinical models or Phase I clinical trials implementing exosomes or other nanocarriers to attack metastatic cancer cells are highlighted.
Collapse
Affiliation(s)
- Ranvir Bhatia
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Joanna Chang
- Department of Biological Sciences, University of Maryland, Baltimore, MD 21250, USA
| | - Jessian L Munoz
- Division of Perinatal Surgery, Texas Children's Hospital, Houston, TX 77030, USA
- Division of Maternal Fetal Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Nykia D Walker
- Department of Biological Sciences, University of Maryland, Baltimore, MD 21250, USA
| |
Collapse
|
2
|
CXCR4/CX43 Regulate Diabetic Neuropathic Pain via Intercellular Interactions between Activated Neurons and Dysfunctional Astrocytes during Late Phase of Diabetes in Rats and the Effects of Antioxidant N-Acetyl-L-Cysteine. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8547563. [PMID: 35799894 PMCID: PMC9256426 DOI: 10.1155/2022/8547563] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 06/15/2022] [Indexed: 12/12/2022]
Abstract
Growing evidence suggests that the interactions between astrocytes and neurons exert important functions in the central sensitization of the spinal cord dorsal horn in rodents with diabetes and neuropathic pain (DNP). However, it still remains unclear how signal transmission occurs in the spinal cord dorsal horn between astrocytes and neurons, especially in subjects with DNP. Chemokine CXC receptor 4 (CXCR4) plays critical roles in DNP, and connexin 43 (CX43), which is also primarily expressed by astrocytes, contributes to the development of neuropathy. We thus postulated that astrocytic and neuronal CXCR4 induces and produces inflammatory factors under persistent peripheral noxious stimulation in DNP, while intercellular CX43 can transmit inflammatory stimulation signals. The results showed that streptozotocin-induced type 1 diabetic rats developed heat hyperalgesia and mechanical allodynia. Diabetes led to persistent neuropathic pain. Diabetic rats developed peripheral sensitization at the early phase (2 weeks) and central sensitization at the late phase (5 weeks) after diabetes induction. Both CXCR4 and CX43, which are localized and coexpressed in neurons and astrocytes, were enhanced significantly in the dorsal horn of spinal cord in rats undergoing DNP during late phase of diabetes, and the CXCR4 antagonist AMD3100 reduced the expression of CX43. The nociceptive behavior was reversed, respectively, by AMD3100 at the early phase and by the antioxidant N-acetyl-L-cysteine (NAC) at the late phase. Furthermore, rats with DNP demonstrated downregulation of glial fibrillary acidic protein (GFAP) as well as upregulation of c-fos in the spinal cord dorsal horn at the late phase compared to the controls, and upregulation of GFAP and downregulation of c-fos were observed upon treatment with NAC. Given that GFAP and c-fos are, respectively, makers of astrocyte and neuronal activation, our findings suggest that CXCR4 as an inflammatory stimulation protein and CX43 as an intercellular signal transmission protein both may induce neurons excitability and astrocytes dysfunction in developing DNP.
Collapse
|
3
|
Differential Expression of BOC, SPOCK2, and GJD3 Is Associated with Brain Metastasis of ER-Negative Breast Cancers. Cancers (Basel) 2021; 13:cancers13122982. [PMID: 34203581 PMCID: PMC8232218 DOI: 10.3390/cancers13122982] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/02/2021] [Accepted: 06/03/2021] [Indexed: 01/05/2023] Open
Abstract
Simple Summary Brain metastasis is diagnosed in 30–50% of metastatic breast cancer patients with currently limited treatment strategies and usually short survival rates. In the present study, we aim to identify genes specifically associated with the development of brain metastasis in breast cancer. Therefore, we compared RNA expression profiles from two groups of patients with metastatic breast cancer, with and without brain involvement. Three genes BOC, SPOCK2, and GJD3 were overexpressed in the group of primary breast cancers which developed brain metastasis. Expression profiles were confirmed in an independent breast cancer cohort for both BOC and SPOCK2. In addition, differential overexpression of SPOCK2 and GJD3 mRNA levels were found to be associated with the development of brain metastasis in an external online database of 204 primary breast cancers. Verification of these genes as biomarkers for brain metastasis development in primary breast cancer is warranted. Abstract Background: Brain metastasis is considered one of the major causes of mortality in breast cancer patients. To invade the brain, tumor cells need to pass the blood-brain barrier by mechanisms that are partially understood. In primary ER-negative breast cancers that developed brain metastases, we found that some of the differentially expressed genes play roles in the T cell response. The present study aimed to identify genes involved in the formation of brain metastasis independently from the T cell response. Method: Previously profiled primary breast cancer samples were reanalyzed. Genes that were found to be differentially expressed were confirmed by RT-PCR and by immunohistochemistry using an independent cohort of samples. Results: BOC, SPOCK2, and GJD3 were overexpressed in the primary breast tumors that developed brain metastasis. BOC expression was successfully validated at the protein level. SPOCK2 was validated at both mRNA and protein levels. SPOCK2 and GJD3 mRNA overexpression were also found to be associated with cerebral metastasis in an external online database consisting of 204 primary breast cancers. Conclusion: The overexpression of BOC, SPOCK2, and GJD3 is associated with the invasion of breast cancer into the brain. Further studies to determine their specific function and potential value as brain metastasis biomarkers are required.
Collapse
|
4
|
Ferrer AI, Trinidad JR, Sandiford O, Etchegaray JP, Rameshwar P. Epigenetic dynamics in cancer stem cell dormancy. Cancer Metastasis Rev 2021; 39:721-738. [PMID: 32394305 DOI: 10.1007/s10555-020-09882-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cancer remains one of the most challenging diseases despite significant advances of early diagnosis and therapeutic treatments. Cancerous tumors are composed of various cell types including cancer stem cells capable of self-renewal, proliferation, differentiation, and invasion of distal tumor sites. Most notably, these cells can enter a dormant cellular state that is resistant to conventional therapies. Thereby, cancer stem cells have the intrinsic potential for tumor initiation, tumor growth, metastasis, and tumor relapse after therapy. Both genetic and epigenetic alterations are attributed to the formation of multiple tumor types. This review is focused on how epigenetic dynamics involving DNA methylation and DNA oxidations are implicated in breast cancer and glioblastoma multiforme. The emergence and progression of these cancer types rely on cancer stem cells with the capacity to enter quiescence also known as a dormant cellular state, which dictates the distinct tumorigenic aggressiveness between breast cancer and glioblastomas.
Collapse
Affiliation(s)
- Alejandra I Ferrer
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
| | - Jonathan R Trinidad
- Department of Biological Sciences, Rutgers University, Newark, NJ, 07102, USA
| | - Oleta Sandiford
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
| | | | - Pranela Rameshwar
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA.
| |
Collapse
|
5
|
Sinha G, Ferrer AI, Moore CA, Naaldijk Y, Rameshwar P. Gap Junctions and Breast Cancer Dormancy. Trends Cancer 2020; 6:348-357. [PMID: 32209448 DOI: 10.1016/j.trecan.2020.01.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 01/22/2020] [Accepted: 01/23/2020] [Indexed: 12/21/2022]
Abstract
Breast cancer (BC) relapse, despite clinical advancement, remains one of the biggest issues in the field. Intercellular communication, specifically via connexin (Cx)-mediated gap junctions (GJs), play a key role in the long-term survival of these, treatment-resistant breast cancer stem cells (CSCs), allowing for relapse. Both basic and clinical evidence reveal dual roles for GJs, in tumor suppression, generally referred to as dormancy, and progression and metastasis. GJ intercellular communication (GJIC) can be mediated by multiple types of Cxs, depending on the organ to which the BC cells metastasize. This review expands on the differential expression of Cx-mediated GJIC between CSCs and niche cells within a given microenvironment.
Collapse
Affiliation(s)
- Garima Sinha
- Rutgers School of Graduate Studies at New Jersey Medical School, Newark, NJ, USA; Department of Medicine - Hematology/Oncology, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Alejandra I Ferrer
- Rutgers School of Graduate Studies at New Jersey Medical School, Newark, NJ, USA; Department of Medicine - Hematology/Oncology, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Caitlyn A Moore
- Rutgers School of Graduate Studies at New Jersey Medical School, Newark, NJ, USA; Department of Medicine - Hematology/Oncology, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Yahaira Naaldijk
- Rutgers School of Graduate Studies at New Jersey Medical School, Newark, NJ, USA
| | - Pranela Rameshwar
- Department of Medicine - Hematology/Oncology, Rutgers New Jersey Medical School, Newark, NJ, USA.
| |
Collapse
|
6
|
Yang M, Zeng C, Li P, Qian L, Ding B, Huang L, Li G, Jiang H, Gong N, Wu W. Impact of CXCR4 and CXCR7 knockout by CRISPR/Cas9 on the function of triple-negative breast cancer cells. Onco Targets Ther 2019; 12:3849-3858. [PMID: 31190884 PMCID: PMC6527053 DOI: 10.2147/ott.s195661] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Background Breast cancer is one of the most common malignancies threatening women's health. Triple-negative breast cancer (TNBC) is a special type of breast cancer with high invasion and metastasis. CXCL12 and its receptors CXCR4 and CXCR7 play a crucial role in the progress of breast cancer. The aim of this study was to investigate the effect of CXCR4 and CXCR7 on the function of TNBC. Materials and methods We used the CRISPR/Cas9 technique to carry out a single knockout of the CXCR4 or CXCR7 gene and co-knockout of CXCR4 and CXCR7 genes in the TNBC cell line (MDA-MB-231). The single knockout and co-knockout cells were screened and verified by PCR sequencing and Western blot assay, the effect of single knockout and co-knockout on the proliferation of TNBC cells was examined using the Cell Counting Kit-8 and colony formation assays, the migration and invasion of TNBC cells were examined by the transwell and wound-healing assays, the changes in the cell cycle distribution after knockout were detected by flow cytometry, and the difference in the migration and invasion of single knockout and co-knockout induced by CXCL12 was observed by adding CXCL12 in the experimental group. Results The single knockout of the CXCR4 or CXCR7 gene significantly reduced the cell proliferation, growth, migration, and invasion and delayed the conversion of the G1/S cycle, while the co-knockout inhibited these biological abilities more significantly. In both the knockout and control groups, the migration and invasion of CXCL12-added cells were significantly stronger than those of the non-CXCL12-added cells, and CXCL12 induced lesser migration and invasion in the CXCR4 and CXCR7 co-knockout group than in the single knockout groups. Conclusion The knockout of the CXCR4 and CXCR7 genes affects the binding capacity and functions of CXCL12, inhibits the malignant progression of TNBC cells significantly, and may become a potential target for the treatment of TNBC.
Collapse
Affiliation(s)
- Meng Yang
- Department of Breast Thyroid Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, People's Republic of China,
| | - Chen Zeng
- Department of Breast Thyroid Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, People's Republic of China,
| | - Peiting Li
- Department of Breast Thyroid Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, People's Republic of China,
| | - Liyuan Qian
- Department of Breast Thyroid Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, People's Republic of China,
| | - Boni Ding
- Department of Breast Thyroid Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, People's Republic of China,
| | - Lihua Huang
- Central Laboratory, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, People's Republic of China
| | - Gang Li
- Department of Breast Thyroid Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, People's Republic of China,
| | - Han Jiang
- Department of Breast Thyroid Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, People's Republic of China,
| | - Ni Gong
- Department of Gastrointestinal Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan Province, People's Republic of China,
| | - Wei Wu
- Department of Breast Thyroid Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, People's Republic of China,
| |
Collapse
|
7
|
Pilarczyk G, Papenfuß F, Bestvater F, Hausmann M. Spatial Arrangements of Connexin43 in Cancer Related Cells and Re-Arrangements under Treatment Conditions: Investigations on the Nano-Scale by Super-Resolution Localization Light Microscopy. Cancers (Basel) 2019; 11:cancers11030301. [PMID: 30836676 PMCID: PMC6468626 DOI: 10.3390/cancers11030301] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 02/25/2019] [Accepted: 02/27/2019] [Indexed: 12/14/2022] Open
Abstract
Cancer studies suggest that the spatial localization of connexin43 (Cx43) could play an important role during tumor genesis and the formation of metastasis. Cx43 has been shown to be upregulated in cancer cells; thereby a shift from Cx43 normal localization in gap junctions in the cell membrane towards a primarily cytoplasmic localization was observed in many studies. So far neither the spatial arrangements of Cx43 in breast cancer cells nor the effects of treatment outcome (ionizing radiation and antibody therapy) on the spatial arrangements of Cx43, have been microscopically studied on the nanoscale. This has brought up the idea to study the micro- and nanoscaled spatial Cx43 arrangements in a model of breast cancer-related cell types, i.e., SkBr3 breast cancer cells, BJ fibroblasts, and primary human internal mammary artery endothelial cells (HIMAECs). The cells were treated with neuregulin1 (NRG1), trastuzumab (Herceptin), or 6MeV-photon irradiation at a dose of 4 Gy. NRG1 stimulates further NRG1 release in the tumor endothelium that may lead to an enhanced tumor protective effect whereas Herceptin, used in antibody treatment, works in an antagonistic fashion to NRG1. After fluorescent labelling with specific antibodies, the molecular positions of Cx43 in the perinuclear cytosol and in the cell periphery at the membrane were determined for the three treatment related applications (NRG1, trastuzumab, 4 Gy irradiation) using confocal laser scanning microscopy (CLSM) and single molecule localization microscopy (SMLM). These techniques enable investigations of Cx43 enrichment and topological arrangements of Cx43 molecules from the micro-scale of a whole cell to the nano-scale of single molecules. In SkBr3 cells with and without radiation treatment high density accumulations were detected which seem to be diluted after NRG1 and trastuzumab treatment although the SMLM distance frequency distributions did not significantly vary. In BJ fibroblasts and HIMAECs differences between periphery and perinuclear cytosol were observed after the different treatment processes. HIMAECs showed significant Cx43 accumulation after NRG1, trastuzumab, and radiation treatment in the perinuclear region whereas in the periphery radiation has less influence as compared to the control. BJ cells were reacting to the treatments by Cx43 accumulations in the perinuclear region but also in the periphery. In conclusion, it was shown that by using CLSM and super-resolution SMLM, treatment effects on the spatial and thus functional arrangements of Cx43 became detectable for investigations of tumor response mechanisms.
Collapse
Affiliation(s)
- Götz Pilarczyk
- Kirchhoff-Institute for Physics, University of Heidelberg, Im Neuenheimer Feld 227, 69120 Heidelberg, Germany.
- Department of High Content Analysis of the Cell "HiCell", BioQuant, University of Heidelberg, Im Neuenheimer Feld 267, 69120 Heidelberg, Germany.
| | - Franziska Papenfuß
- Kirchhoff-Institute for Physics, University of Heidelberg, Im Neuenheimer Feld 227, 69120 Heidelberg, Germany.
| | - Felix Bestvater
- Core Facility Unit Light Microscopy, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany.
| | - Michael Hausmann
- Kirchhoff-Institute for Physics, University of Heidelberg, Im Neuenheimer Feld 227, 69120 Heidelberg, Germany.
| |
Collapse
|
8
|
Munoz JL, Walker ND, Mareedu S, Pamarthi SH, Sinha G, Greco SJ, Rameshwar P. Cycling Quiescence in Temozolomide Resistant Glioblastoma Cells Is Partly Explained by microRNA-93 and -193-Mediated Decrease of Cyclin D. Front Pharmacol 2019; 10:134. [PMID: 30853911 PMCID: PMC6395452 DOI: 10.3389/fphar.2019.00134] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 02/05/2019] [Indexed: 12/30/2022] Open
Abstract
Glioblastoma multiforme (GBM) is a fatal malignancy of the central nervous system, commonly associated with chemoresistance. The alkylating agent Temozolomide (TMZ) is the front-line chemotherapeutic agent and has undergone intense studies on resistance. These studies reported on mismatch repair gene upregulation, ABC-targeted drug efflux, and cell cycle alterations. The mechanism by which TMZ induces cell cycle arrest has not been well-established. TMZ-resistant GBM cells have been linked to microRNA (miRNA) and exosomes. A cell cycle miRNA array identified distinct miRNAs only in exosomes from TMZ-resistant GBM cell lines and primary spheres. We narrowed the miRs to miR-93 and -193 and showed in computational analyses that they could target Cyclin D1. Since Cyclin D1 is a major regulator of cell cycle progression, we performed cause-effect studies and showed a blunting effects of miR-93 and -193 in Cyclin D1 expression. These two miRs also decreased cell cycling quiescence and induced resistance to TMZ. Taken together, our data provide a mechanism by which GBM cells can exhibit TMZ-induced resistance through miRNA targeting of Cyclin D1. The data provide a number of therapeutic approaches to reverse chemoresistance at the miRNA, exosomal and cell cycle points.
Collapse
Affiliation(s)
- Jessian L Munoz
- Rutgers New Jersey Medical School, Rutgers University, Newark, NJ, United States.,Rutgers School of Graduate Studies at New Jersey Medical School, Rutgers University, Newark, NJ, United States
| | - Nykia D Walker
- Rutgers School of Graduate Studies at New Jersey Medical School, Rutgers University, Newark, NJ, United States
| | - Satvik Mareedu
- Rutgers New Jersey Medical School, Rutgers University, Newark, NJ, United States.,Rutgers School of Graduate Studies at New Jersey Medical School, Rutgers University, Newark, NJ, United States
| | - Sri Harika Pamarthi
- Rutgers New Jersey Medical School, Rutgers University, Newark, NJ, United States
| | - Garima Sinha
- Rutgers New Jersey Medical School, Rutgers University, Newark, NJ, United States.,Rutgers School of Graduate Studies at New Jersey Medical School, Rutgers University, Newark, NJ, United States
| | - Steven J Greco
- Rutgers New Jersey Medical School, Rutgers University, Newark, NJ, United States
| | - Pranela Rameshwar
- Rutgers New Jersey Medical School, Rutgers University, Newark, NJ, United States.,Rutgers School of Graduate Studies at New Jersey Medical School, Rutgers University, Newark, NJ, United States
| |
Collapse
|
9
|
The Role of Connexin-43 in the Inflammatory Process: A New Potential Therapy to Influence Keratitis. J Ophthalmol 2019; 2019:9312827. [PMID: 30805212 PMCID: PMC6360563 DOI: 10.1155/2019/9312827] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 11/12/2018] [Accepted: 11/19/2018] [Indexed: 12/22/2022] Open
Abstract
The studies outlined in this review highlight the relationship between inflammatory signaling molecules and connexin-43 (Cx43). Gap junction (GJ) channels and hemichannels (HCs) participate in the metabolic activity between intra- and extracellular space. Some ions and small molecules are exchanged from cell to cell or cell to extracellular space to affect the process of inflammation via GJ. We analyzed the effects of signaling molecules, such as innate immunity messengers, transcription factors, LPS, cytokine, inflammatory chemokines, and MMPs, on Cx43 expression during the inflammatory process. At the same time, we found that these signaling molecules play a critical role in the pathogenesis of keratitis. Thus, we assessed the function of Cx43 during inflammatory corneal disease. Corneal healing plays an essential role in the late stage of keratitis. We found that Cx43 is involved in wound healing. Studies have shown that the decrease of Cx43 can decrease the time of healing. We also report several Cx43 mimic peptides which can inhibit the activity of Cx43 Hc to mediate the releasing of adenosine triphosphate (ATP), which may in turn influence the inflammatory process.
Collapse
|
10
|
Eltoukhy HS, Sinha G, Moore CA, Gergues M, Rameshwar P. Secretome within the bone marrow microenvironment: A basis for mesenchymal stem cell treatment and role in cancer dormancy. Biochimie 2018; 155:92-103. [PMID: 29859990 DOI: 10.1016/j.biochi.2018.05.018] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 05/29/2018] [Indexed: 02/07/2023]
Abstract
The secretome produced by cells within the bone marrow is significant to homeostasis. The bone marrow, a well-studied organ, has multiple niches with distinct roles for supporting stem cell functions. Thus, an understanding of mediators involved in the regulation of stem cells could serve as a model for clinical problems and solutions such as tissue repair and regeneration. The exosome secretome of bone marrow stem cells is a developing area of research with respect to the regenerative potential by bone marrow cell, particularly the mesenchymal stem cells. The bone marrow niche regulates endogenous processes such as hematopoiesis but could also support the survival of tumors such as facilitating the cancer stem cells to exist in dormancy for decades. The bone marrow-derived secretome will be critical to future development of therapeutic strategies for oncologic diseases, in addition to regenerative medicine. This article discusses the importance for parallel studies to determine how the same secretome may compromise safety during the use of stem cells in regenerative medicine.
Collapse
Affiliation(s)
- Hussam S Eltoukhy
- Department of Medicine - Division of Hematology/Oncology, Rutgers, New Jersey Medical School, Newark, NJ 07103, USA
| | - Garima Sinha
- Department of Medicine - Division of Hematology/Oncology, Rutgers, New Jersey Medical School, Newark, NJ 07103, USA
| | - Caitlyn A Moore
- Department of Medicine - Division of Hematology/Oncology, Rutgers, New Jersey Medical School, Newark, NJ 07103, USA
| | - Marina Gergues
- Department of Medicine - Division of Hematology/Oncology, Rutgers, New Jersey Medical School, Newark, NJ 07103, USA
| | - Pranela Rameshwar
- Department of Medicine - Division of Hematology/Oncology, Rutgers, New Jersey Medical School, Newark, NJ 07103, USA.
| |
Collapse
|
11
|
Wang S, Zhang S, Zhao Z, Zhang C, Yang X, Wang Y. Connexin 43 enhances paclitaxel cytotoxicity in colorectal cancer cell lines. Exp Ther Med 2017; 14:1212-1218. [PMID: 28810580 PMCID: PMC5526126 DOI: 10.3892/etm.2017.4589] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Accepted: 04/07/2017] [Indexed: 01/08/2023] Open
Abstract
Colorectal cancer has a relatively low sensitivity to paclitaxel. The purpose of this study was to investigate the role of connexin 43 (Cx43), which is a structural component of gap junctional communication (GJC), in paclitaxel cytotoxicity in colorectal cancer cells. Three colorectal cancer cell lines (HCT106, HCT116 and LoVo) were transfected with Cx43 and used to examine paclitaxel cytotoxicity. A western blot assay was used to confirm Cx43 expression in transfected cell lines as well as the expression of several proteins that are associated with paclitaxel cytotoxicity. A parachute dye-coupling assay was used to measure GJC function. An MTT assay was used to analyze the viability of paclitaxel-treated cells. Cx43 expression level and GJC function were significantly upregulated by the transfection (P<0.05). The viability of transfected cells was significantly inhibited compared with that of untransfected cells when treated with paclitaxel (20 or 80 nM) at high culture density but not at low culture density (P<0.05). Cx43 transfection significantly increased the mitotic arrest, tubulin polymerization and apoptosis effects of paclitaxel (P<0.05). It was also found that paclitaxel had an inhibitory effect on GJC function after 12 h of treatment in LoVo cells (P<0.05). These results indicate that Cx43 may serve as a target of paclitaxel chemotherapy for colorectal cancer.
Collapse
Affiliation(s)
- Siqi Wang
- Laboratory of Biomedicine and Nanophotonics, Tianjin Union Medical Center, Tianjin 300121, P.R. China
| | - Shiwu Zhang
- Department of Pathology, Tianjin Union Medical Center, Tianjin 300121, P.R. China
| | - Zhenying Zhao
- Department of Pharmacy, Tianjin Union Medical Center, Tianjin 300121, P.R. China
| | - Chunze Zhang
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin 300121, P.R. China
| | - Xiaoyun Yang
- Department of Pathology, Tianjin Union Medical Center, Tianjin 300121, P.R. China
| | - Yijia Wang
- Laboratory of Biomedicine and Nanophotonics, Tianjin Union Medical Center, Tianjin 300121, P.R. China
| |
Collapse
|
12
|
Non-coding RNA as mediators in microenvironment–breast cancer cell communication. Cancer Lett 2016; 380:289-95. [PMID: 26582656 DOI: 10.1016/j.canlet.2015.11.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Revised: 11/04/2015] [Accepted: 11/06/2015] [Indexed: 12/18/2022]
Abstract
The tumor microenvironment has a critical role in the survival and decision of the cancer cells. These include support by enhanced angiogenesis, and metastasis or adaptation of dormancy. This article discusses methods by which the microenvironment sustains the tumor. This process is important as it will identify avenues of drug targets. Non-coding RNAs (ncRNAs) are evolving as key mediators in the interaction between the cancer cells and the microenvironment. Thus, the question is how to develop methods to effectively block the effects of the ncRNA and/or to introduce them to prevent metastasis, dormancy or to reverse dormancy. We focused on the advantages of using mesenchymal stem cells (MSCs) for RNA delivery. MSCs can be available as "off-the-shelf" cells. Thus far, MSCs are shown to be safe when transplanted across allogeneic barriers. We discussed the various methods by which MSCs can interact with cancer cells to deliver ncRNA or antagomirs. We also include the advances and possible confounds of using these methods. Overall, this review article provides a potential method by which MSCs can be used for effective delivery of nucleic acid to treat cancer.
Collapse
|
13
|
Temozolomide resistance in glioblastoma occurs by miRNA-9-targeted PTCH1, independent of sonic hedgehog level. Oncotarget 2015; 6:1190-201. [PMID: 25595896 PMCID: PMC4359226 DOI: 10.18632/oncotarget.2778] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2014] [Accepted: 11/19/2014] [Indexed: 12/30/2022] Open
Abstract
Glioblastoma Multiforme (GBM), the most common and lethal adult primary tumor of the brain, showed a link between Sonic Hedgehog (SHH) pathway in the resistance to temozolomide (TMZ). PTCH1, the SHH receptor, can tonically represses signaling by endocytosis. We asked how the decrease in PTCH1 in GBM cells could lead to TMZ-resistance. TMZ resistant GBM cells have increased PTCH1 mRNA and reduced protein. Knockdown of Dicer, a Type III RNAase, indicated that miRNAs can explain the decreased PTCH1 in TMZ resistant cells. Computational studies, real-time PCR, reporter gene studies, western blots, target protector oligos and ectopic expression identified miR-9 as the target of PTCH1 in resistant GBM cells with concomitant activation of SHH signaling. MiR-9 mediated increases in the drug efflux transporters, MDR1 and ABCG2. MiR-9 was increased in the tissues from GBM patients and in an early passage GBM cell line from a patient with recurrent GBM but not from a naïve patient. Pharmacological inhibition of SHH signaling sensitized the GBM cells to TMZ. Taken together, miR-9 targets PTCH1 in GBM cells by a SHH-independent method in GBM cells for TMZ resistance. The identified pathways could lead to new strategies to target GBM with combinations of drugs.
Collapse
|
14
|
Walker ND, Patel J, Munoz JL, Hu M, Guiro K, Sinha G, Rameshwar P. The bone marrow niche in support of breast cancer dormancy. Cancer Lett 2015; 380:263-71. [PMID: 26546045 DOI: 10.1016/j.canlet.2015.10.033] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 10/13/2015] [Accepted: 10/27/2015] [Indexed: 12/15/2022]
Abstract
Despite the success in detecting breast cancer (BC) early and, with aggressive therapeutic intervention, BC remains a clinical problem. The bone marrow (BM) is a favorable metastatic site for breast cancer cells (BCCs). In BM, the survival of BCCs is partly achieved by the supporting microenvironment, including the presence of immune suppressive cells such as mesenchymal stem cells (MSCs). The heterogeneity of BCCs brings up the question of how each subset interacts with the BM microenvironment. The cancer stem cells (CSCs) survive in the BM as cycling quiescence cells and, forming gap junctional intercellular communication (GJIC) with the hematopoietic supporting stromal cells and MSCs. This type of communication has been identified close to the endosteum. Additionally, dormancy can occur by soluble mediators such as cytokines and also by the exchange of exosomes. These latter mechanisms are reviewed in the context of metastasis of BC to the BM for transition as dormant cells. The article also discusses how immune cells such as macrophages and regulatory T-cells facilitate BC dormancy. The challenges of studying BC dormancy in 2-dimensional (2-D) system are also incorporated by proposing 3-D system by engineering methods to recapitulate the BM microenvironment.
Collapse
Affiliation(s)
- Nykia D Walker
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA; Graduate School of Biomedical Sciences at New Jersey Medical School, Newark, NJ, USA
| | - Jimmy Patel
- Graduate School of Biomedical Sciences at New Jersey Medical School, Newark, NJ, USA
| | - Jessian L Munoz
- Ob/Gyn and Women's Health Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Madeleine Hu
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA; Graduate School of Biomedical Sciences at New Jersey Medical School, Newark, NJ, USA
| | - Khadidiatou Guiro
- Graduate School of Biomedical Sciences at New Jersey Medical School, Newark, NJ, USA
| | - Garima Sinha
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA; Graduate School of Biomedical Sciences at New Jersey Medical School, Newark, NJ, USA
| | - Pranela Rameshwar
- Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA; Graduate School of Biomedical Sciences at New Jersey Medical School, Newark, NJ, USA.
| |
Collapse
|
15
|
Munoz JL, Rodriguez-Cruz V, Greco SJ, Nagula V, Scotto KW, Rameshwar P. Temozolomide Induces the Production of Epidermal Growth Factor to Regulate MDR1 Expression in Glioblastoma Cells. Mol Cancer Ther 2014; 13:2399-411. [DOI: 10.1158/1535-7163.mct-14-0011] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
16
|
Patel SA, Dave MA, Bliss SA, Giec-Ujda AB, Bryan M, Pliner LF, Rameshwar P. T reg/Th17 polarization by distinct subsets of breast cancer cells is dictated by the interaction with mesenchymal stem cells. JOURNAL OF CANCER STEM CELL RESEARCH 2014; 2014:e1003. [PMID: 25705705 PMCID: PMC4334154 DOI: 10.14343/jcscr.2014.2e1003] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Breast cancer (BC) cells (BCCs) exist within a hierarchy beginning with cancer stem cells (CSCs). Unsorted BCCs interact with mesenchymal stem cells (MSCs) to induce regulatory T cells (Tregs). This study investigated how distinct BCC subsets interacted with MSCs to polarize T-cell response, Tregs versus T helper 17 (Th17). This study tested BC initiating cells (CSCs) and the relatively more mature early and late BC progenitors. CSCs interacted with the highest avidity to MSCs. This interaction required CXCR4 and connexin 43 (Cx43)-dependant gap junctional intercellular communication (GJIC). This interaction induced Treg whereas interactions between MSCs and the progenitors induced Th17 response. The increases in Treg and Th17 depended on MSCs but not CTLA-4, which was increased in the presence of MSCs. Studies with BM stroma (fibroblasts) and MSCs from the same donors, indicated specific effects of MSCs. In total, MSC-CSC interaction required CXCR4 for GJIC. This led to increased Tregs and TGFβ, and decreased Th17. In contrast, late and early BCCs showed reduced formation of GJIC, decreased Treg and increased Th17 and IL-17. These findings have significance to the methods by which CSCs evade the immune response. The findings could provide methods of intervention to reverse immune-mediated protection and support of BC.
Collapse
Affiliation(s)
- Shyam A. Patel
- Dept of Medicine, Hematology/Oncology, New Jersey Medical School, Rutgers School of Biomedical Health Sciences, Newark, NJ, USA
- Graduate School of Biomedical Sciences, Rutgers School of Biomedical Health Sciences, Newark, NJ, USA
| | - Meneka A. Dave
- Dept of Medicine, Hematology/Oncology, New Jersey Medical School, Rutgers School of Biomedical Health Sciences, Newark, NJ, USA
| | - Sarah A. Bliss
- Dept of Medicine, Hematology/Oncology, New Jersey Medical School, Rutgers School of Biomedical Health Sciences, Newark, NJ, USA
- Graduate School of Biomedical Sciences, Rutgers School of Biomedical Health Sciences, Newark, NJ, USA
| | - Agata B. Giec-Ujda
- Dept of Medicine, Hematology/Oncology, New Jersey Medical School, Rutgers School of Biomedical Health Sciences, Newark, NJ, USA
- Graduate School of Biomedical Sciences, Rutgers School of Biomedical Health Sciences, Newark, NJ, USA
| | - Margarette Bryan
- Dept of Medicine, Hematology/Oncology, New Jersey Medical School, Rutgers School of Biomedical Health Sciences, Newark, NJ, USA
| | - Lillian F. Pliner
- Dept of Medicine, Hematology/Oncology, New Jersey Medical School, Rutgers School of Biomedical Health Sciences, Newark, NJ, USA
| | - Pranela Rameshwar
- Dept of Medicine, Hematology/Oncology, New Jersey Medical School, Rutgers School of Biomedical Health Sciences, Newark, NJ, USA
| |
Collapse
|
17
|
Bliss SA, Greco SJ, Rameshwar P. Hierarchy of breast cancer cells: key to reverse dormancy for therapeutic intervention. Stem Cells Transl Med 2014; 3:782-6. [PMID: 24833590 DOI: 10.5966/sctm.2014-0013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
An understanding of how cancer cells adapt dormancy would allow for targeted treatment. The current literature suggests that the cancer stem cells might be the major cells with the ability to become quiescent and to resist current drug treatment. The properties of cancer stem cells and healthy stem cells are functionally similar, thereby posing a challenge to target the dormant cells. The bone marrow is particularly a challenge because the dormant breast cancer cells are close to the endosteum, which is also home to the endogenous hematopoietic stem cells. Here we discuss how research studies could bring an understanding of the cellular and molecular interactions between the cancer stem cells and cells within the bone marrow microenvironment. This will allow for intervention to reverse dormancy for targeted treatment. The treatment will require studies within the normal organ functions to ensure treatment without toxicity.
Collapse
Affiliation(s)
- Sarah A Bliss
- Department of Medicine, Hematology/Oncology, New Jersey Medical School and Graduate School of Biomedical Sciences, Rutgers University, Newark, New Jersey, USA
| | - Steven J Greco
- Department of Medicine, Hematology/Oncology, New Jersey Medical School and Graduate School of Biomedical Sciences, Rutgers University, Newark, New Jersey, USA
| | - Pranela Rameshwar
- Department of Medicine, Hematology/Oncology, New Jersey Medical School and Graduate School of Biomedical Sciences, Rutgers University, Newark, New Jersey, USA
| |
Collapse
|
18
|
Temozolomide resistance in glioblastoma cells occurs partly through epidermal growth factor receptor-mediated induction of connexin 43. Cell Death Dis 2014; 5:e1145. [PMID: 24675463 PMCID: PMC3973225 DOI: 10.1038/cddis.2014.111] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 02/07/2014] [Accepted: 02/17/2014] [Indexed: 12/20/2022]
Abstract
Glioblastoma Multiforme (GBM) is an aggressive adult primary brain tumor with poor prognosis. GBM patients develop resistance to the frontline chemotherapy, temozolomide (TMZ). As the connexins (Cx) have been shown to have a complex role in GBM, we investigated the role of Cx43 in TMZ resistance. Cx43 was increased in the TMZ-resistant low passage and cell lines. This correlated with the data in The Cancer Genome Atlas. Cx43 knockdown, reporter gene assays, chromatin immunoprecipitation assay, real-time PCR and western blots verified a role for Cx43 in TMZ resistance. This occurred by TMZ-resistant GBM cells being able to activate epidermal growth factor receptor (EGFR). In turn, EGFR activated the JNK-ERK1/2-AP-1 axis to induce Cx43. The increased Cx43 was functional as indicated by gap junctional intercellular communication among the resistant GBM cells. Cell therapy could be a potential method to deliver drugs, such as anti-EGF to tumor cells. Similar strategies could be used to reverse the expression of Cx43 to sensitize GBM cells to TMZ. The studies showed the potential for targeting EGF in immune therapy. These agents can be used in conjunction with stem cell therapy to treat GBM.
Collapse
|
19
|
Delivery of Functional Anti-miR-9 by Mesenchymal Stem Cell-derived Exosomes to Glioblastoma Multiforme Cells Conferred Chemosensitivity. MOLECULAR THERAPY. NUCLEIC ACIDS 2013; 2:e126. [PMID: 24084846 PMCID: PMC4027430 DOI: 10.1038/mtna.2013.60] [Citation(s) in RCA: 398] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Accepted: 08/15/2013] [Indexed: 12/21/2022]
Abstract
Glioblastoma multiforme (GBM), the most common and lethal tumor of the adult brain, generally shows chemo- and radioresistance. MicroRNAs (miRs) regulate physiological processes, such as resistance of GBM cells to temozolomide (TMZ). Although miRs are attractive targets for cancer therapeutics, the effectiveness of this approach requires targeted delivery. Mesenchymal stem cells (MSCs) can migrate to the sites of cancers, including GBM. We report on an increase in miR-9 in TMZ-resistant GBM cells. miR-9 was involved in the expression of the drug efflux transporter, P-glycoprotein. To block miR-9, methods were developed with Cy5-tagged anti-miR-9. Dye-transfer studies indicated intracellular communication between GBM cells and MSCs. This occurred by gap junctional intercellular communication and the release of microvesicles. In both cases, anti-miR-9 was transferred from MSCs to GBM cells. However, the major form of transfer occurred with the microvesicles. The delivery of anti-miR-9 to the resistant GBM cells reversed the expression of the multidrug transporter and sensitized the GBM cells to TMZ, as shown by increased cell death and caspase activity. The data showed a potential role for MSCs in the functional delivery of synthetic anti-miR-9 to reverse the chemoresistance of GBM cells.
Collapse
|