1
|
Monaco ML, Filpi GA, Kohler SL, Eversole R, Idris OA, Essani K. Oncolytic Tanapoxvirus Recombinants Expressing Flagellin C or Mouse Interleukin-2 Are Capable of Regressing Human Triple-Negative Breast Cancer Xenografts in Immuno-Competent BALB/c Nude Mice. Pathogens 2024; 13:402. [PMID: 38787254 PMCID: PMC11124456 DOI: 10.3390/pathogens13050402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/25/2024] [Accepted: 05/12/2024] [Indexed: 05/25/2024] Open
Abstract
Triple-negative breast cancer (TNBC) in humans is the most aggressive and deadly form of BC. Although TNBCs are about 15 percent of the total number of BC cases, they are associated with the highest mortalities. Current treatment options are limited, and most modalities are toxic and have not increased the 5-year survival rates of TNBC. Many oncolytic viruses are emerging as potential therapies for TNBC. In this study, two Tanapoxvirus (TPV) recombinants, one expressing FliC and the other expressing mouse interleukin-2 (mIL-2), were assessed for their efficacy in an immuno-competent xenograft mouse model. MDA-MB-231 tumors were planted in BALB/c nude mice, treated, made immuno-competent via adoptive transfer of splenocytes from healthy BALB/c donors, and then monitored for 40 days. TPV/Δ2L/66R/FliC and TPV/Δ66R/mIL-2 demonstrated significant tumor reduction (p = 0.01602 and p = 0.03890, respectively) compared to the reconstituted control (RC), whereas wtTPV did not. Pathological analyses of treated tumors revealed cells consistent with lymphocyte and plasma cell morphology in reconstituted mice treated with TPV recombinants. Anti-viral plaque reduction assays conducted using harvested serum from treated animals indicated the presence of anti-TPV antibodies in mice reconstituted and treated with TPV that were missing from immune-deficient nude mice, including those exposed to TPV and of statistically equivalent serum concentrations to normal BALB/c mice immunized against TPV. The results suggest immuno-deficient BALB/c nude mice can become immuno-competent via adoptive transfer of splenocytes from genetically identical donors and allow for testing of tumor xenografts in a competent model system. The TPV recombinants tested should be further studied for the potential treatment of human TNBC.
Collapse
Affiliation(s)
| | | | | | | | | | - Karim Essani
- Laboratory of Virology, Department of Biological Sciences, Western Michigan University, Kalamazoo, MI 49008, USA; (M.L.M.); (G.A.F.); (S.L.K.); (R.E.); (O.A.I.)
| |
Collapse
|
2
|
Ling B, Ye G, Qin C, Liao X, Yang R, Su L, Qi G. IGSF10 inhibits the metastasis of lung adenocarcinoma via the Spi-B/Integrin-β1 signaling pathway. J Biochem Mol Toxicol 2024; 38:e23693. [PMID: 38622980 DOI: 10.1002/jbt.23693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 06/16/2023] [Accepted: 03/12/2024] [Indexed: 04/17/2024]
Abstract
IGSF10, a protein that belongs to the immunoglobulin superfamily, is involved in regulating the early migration of neurons that produce gonadotropin-releasing hormone and performs a fundamental function in development. Our previous study confirmed that the mRNA expression level of IGSF10 may be a protective prognosis factor for lung adenocarcinoma (LUAD) patients. However, the specific mechanisms of IGSF10 are still unclear. In this research, it was shown that the protein level of IGSF10 was down-modulated in LUAD tissues and had a link to the clinical and pathological characteristics as well as the patient's prognosis in LUAD. Importantly, IGSF10 regulates the metastatic ability of LUAD cells in vitro and in vivo. It was proven in a mechanistic sense that IGSF10 inhibits the capacity of LUAD cells to metastasize through the Spi-B/Integrin-β1 signaling pathway. These findings gave credence to the premise that IGSF10 performed a crucial function in LUAD.
Collapse
Affiliation(s)
- Bo Ling
- College of Pharmacy, Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Guangbin Ye
- College of basic Medical Sciences, Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Chunyan Qin
- Department of Pulmonary and Critical Care Medicine, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Xianjiu Liao
- College of Pharmacy, Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Ruirui Yang
- Institute of Life Sciences, Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Lili Su
- Institute of Life Sciences, Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Guangzi Qi
- College of public health and management, Youjiang Medical University for Nationalities, Baise, Guangxi, China
| |
Collapse
|
3
|
Sadri F, Hosseini SF, Rezaei Z, Fereidouni M. Hippo-YAP/TAZ signaling in breast cancer: Reciprocal regulation of microRNAs and implications in precision medicine. Genes Dis 2024; 11:760-771. [PMID: 37692482 PMCID: PMC10491881 DOI: 10.1016/j.gendis.2023.01.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 12/09/2022] [Accepted: 01/29/2023] [Indexed: 09/12/2023] Open
Abstract
Breast cancer is a molecularly heterogeneous disease and the most common female malignancy. In recent years, therapy approaches have evolved to accommodate molecular diversity, with a focus on more biologically based therapies to minimize negative consequences. To regulate cell fate in human breast cells, the Hippo signaling pathway has been associated with the alpha subtype of estrogen receptors. This pathway regulates tissue size, regeneration, and healing, as well as the survival of tissue-specific stem cells, proliferation, and apoptosis in a variety of organs, allowing for cell differentiation. Hippo signaling is mediated by the kinases MST1, MST2, LATS1, and LATS2, as well as the adaptor proteins SAV1 and MOB. These kinases phosphorylate the downstream effectors of the Hippo pathway, yes-associated protein (YAP), and transcriptional coactivator with PDZ-binding motif (TAZ), suppressing the expression of their downstream target genes. The Hippo signaling pathway kinase cascade plays a significant role in all cancers. Understanding the principles of this kinase cascade would prevent the occurrence of breast cancer. In recent years, small noncoding RNAs, or microRNAs, have been implicated in the development of several malignancies, including breast cancer. The interconnections between miRNAs and Hippo signaling pathway core proteins in the breast, on the other hand, remain poorly understood. In this review, we focused on highlighting the Hippo signaling system, its key parts, its importance in breast cancer, and its regulation by miRNAs and other related pathways.
Collapse
Affiliation(s)
- Farzad Sadri
- Student Research Committee, Birjand University of Medical Sciences, Birjand 9717853577, Iran
| | | | - Zohreh Rezaei
- Department of Biology, University of Sistan and Baluchestan, Zahedan 9816745785, Iran
| | - Mohammad Fereidouni
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand 9717853577, Iran
| |
Collapse
|
4
|
Zhou M, Ma Y, Chiang CC, Rock EC, Butler SC, Anne R, Yatsenko S, Gong Y, Chen YC. Single-cell morphological and transcriptome analysis unveil inhibitors of polyploid giant breast cancer cells in vitro. Commun Biol 2023; 6:1301. [PMID: 38129519 PMCID: PMC10739852 DOI: 10.1038/s42003-023-05674-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 12/04/2023] [Indexed: 12/23/2023] Open
Abstract
Considerable evidence suggests that breast cancer therapeutic resistance and relapse can be driven by polyploid giant cancer cells (PGCCs). The number of PGCCs increases with the stages of disease and therapeutic stress. Given the importance of PGCCs, it remains challenging to eradicate them. To discover effective anti-PGCC compounds, there is an unmet need to rapidly distinguish compounds that kill non-PGCCs, PGCCs, or both. Here, we establish a single-cell morphological analysis pipeline with a high throughput and great precision to characterize dynamics of individual cells. In this manner, we screen a library to identify promising compounds that inhibit all cancer cells or only PGCCs (e.g., regulators of HDAC, proteasome, and ferroptosis). Additionally, we perform scRNA-Seq to reveal altered cell cycle, metabolism, and ferroptosis sensitivity in breast PGCCs. The combination of single-cell morphological and molecular investigation reveals promising anti-PGCC strategies for breast cancer treatment and other malignancies.
Collapse
Affiliation(s)
- Mengli Zhou
- UPMC Hillman Cancer Center, University of Pittsburgh, 5115 Centre Ave, Pittsburgh, PA, 15232, USA
- Department of Computational and Systems Biology, University of Pittsburgh, 3420 Forbes Avenue, Pittsburgh, PA, 15260, USA
- Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Yushu Ma
- UPMC Hillman Cancer Center, University of Pittsburgh, 5115 Centre Ave, Pittsburgh, PA, 15232, USA
- Department of Computational and Systems Biology, University of Pittsburgh, 3420 Forbes Avenue, Pittsburgh, PA, 15260, USA
| | - Chun-Cheng Chiang
- UPMC Hillman Cancer Center, University of Pittsburgh, 5115 Centre Ave, Pittsburgh, PA, 15232, USA
- Department of Computational and Systems Biology, University of Pittsburgh, 3420 Forbes Avenue, Pittsburgh, PA, 15260, USA
| | - Edwin C Rock
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, 3700 O'Hara Street, Pittsburgh, PA, 15260, USA
| | - Samuel Charles Butler
- UPMC Hillman Cancer Center, University of Pittsburgh, 5115 Centre Ave, Pittsburgh, PA, 15232, USA
| | - Rajiv Anne
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, 3700 O'Hara Street, Pittsburgh, PA, 15260, USA
| | - Svetlana Yatsenko
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA
- Magee Womens Research Institute, Pittsburgh, PA, USA
| | - Yinan Gong
- UPMC Hillman Cancer Center, University of Pittsburgh, 5115 Centre Ave, Pittsburgh, PA, 15232, USA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Yu-Chih Chen
- UPMC Hillman Cancer Center, University of Pittsburgh, 5115 Centre Ave, Pittsburgh, PA, 15232, USA.
- Department of Computational and Systems Biology, University of Pittsburgh, 3420 Forbes Avenue, Pittsburgh, PA, 15260, USA.
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, 3700 O'Hara Street, Pittsburgh, PA, 15260, USA.
- CMU-Pitt Ph.D. Program in Computational Biology, University of Pittsburgh, 3420 Forbes Avenue, Pittsburgh, PA, 15260, USA.
| |
Collapse
|
5
|
Tariq M, Richard V, Kerin MJ. MicroRNAs as Molecular Biomarkers for the Characterization of Basal-like Breast Tumor Subtype. Biomedicines 2023; 11:3007. [PMID: 38002007 PMCID: PMC10669494 DOI: 10.3390/biomedicines11113007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/03/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
Breast cancer is a heterogeneous disease highlighted by the presence of multiple tumor variants and the basal-like breast cancer (BLBC) is considered to be the most aggressive variant with limited therapeutics and a poor prognosis. Though the absence of detectable protein and hormonal receptors as biomarkers hinders early detection, the integration of genomic and transcriptomic profiling led to the identification of additional variants in BLBC. The high-throughput analysis of tissue-specific micro-ribonucleic acids (microRNAs/miRNAs) that are deemed to have a significant role in the development of breast cancer also displayed distinct expression profiles in each subtype of breast cancer and thus emerged to be a robust approach for the precise characterization of the BLBC subtypes. The classification schematic of breast cancer is still a fluid entity that continues to evolve alongside technological advancement, and the transcriptomic profiling of tissue-specific microRNAs is projected to aid in the substratification and diagnosis of the BLBC tumor subtype. In this review, we summarize the current knowledge on breast tumor classification, aim to collect comprehensive evidence based on the microRNA expression profiles, and explore their potential as prospective biomarkers of BLBC.
Collapse
Affiliation(s)
| | - Vinitha Richard
- Discipline of Surgery, Lambe Institute for Translational Research, H91 TK33 Galway, Ireland;
| | - Michael J. Kerin
- Discipline of Surgery, Lambe Institute for Translational Research, H91 TK33 Galway, Ireland;
| |
Collapse
|
6
|
Hossain F, Ucar DA, Monticone G, Ran Y, Majumder S, Larter K, Luu H, Wyczechowska D, Heidari S, Xu K, Shanthalingam S, Matossian M, Xi Y, Burow M, Collins-Burow B, Del Valle L, Hicks C, Zabaleta J, Golde T, Osborne B, Miele L. Sulindac sulfide as a non-immune suppressive γ-secretase modulator to target triple-negative breast cancer. Front Immunol 2023; 14:1244159. [PMID: 37901240 PMCID: PMC10612326 DOI: 10.3389/fimmu.2023.1244159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 09/18/2023] [Indexed: 10/31/2023] Open
Abstract
Introduction Triple-negative breast cancer (TNBC) comprises a heterogeneous group of clinically aggressive tumors with high risk of recurrence and metastasis. Current pharmacological treatment options remain largely limited to chemotherapy. Despite promising results, the efficacy of immunotherapy and chemo-immunotherapy in TNBC remains limited. There is strong evidence supporting the involvement of Notch signaling in TNBC progression. Expression of Notch1 and its ligand Jagged1 correlate with poor prognosis. Notch inhibitors, including g-secretase inhibitors (GSIs), are quite effective in preclinical models of TNBC. However, the success of GSIs in clinical trials has been limited by their intestinal toxicity and potential for adverse immunological effects, since Notch plays key roles in T-cell activation, including CD8 T-cells in tumors. Our overarching goal is to replace GSIs with agents that lack their systemic toxicity and ideally, do not affect tumor immunity. We identified sulindac sulfide (SS), the active metabolite of FDA-approved NSAID sulindac, as a potential candidate to replace GSIs. Methods We investigated the pharmacological and immunotherapeutic properties of SS in TNBC models in vitro, ex-vivo and in vivo. Results We confirmed that SS, a known γ-secretase modulator (GSM), inhibits Notch1 cleavage in TNBC cells. SS significantly inhibited mammosphere growth in all human and murine TNBC models tested. In a transplantable mouse TNBC tumor model (C0321), SS had remarkable single-agent anti-tumor activity and eliminated Notch1 protein expression in tumors. Importantly, SS did not inhibit Notch cleavage in T- cells, and the anti-tumor effects of SS were significantly enhanced when combined with a-PD1 immunotherapy in our TNBC organoids and in vivo. Discussion Our data support further investigation of SS for the treatment of TNBC, in conjunction with chemo- or -chemo-immunotherapy. Repurposing an FDA-approved, safe agent for the treatment of TNBC may be a cost-effective, rapidly deployable therapeutic option for a patient population in need of more effective therapies.
Collapse
Affiliation(s)
- Fokhrul Hossain
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans (LSUHSC-NO), New Orleans, LA, United States
| | - Deniz A. Ucar
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans (LSUHSC-NO), New Orleans, LA, United States
| | - Giulia Monticone
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans (LSUHSC-NO), New Orleans, LA, United States
| | - Yong Ran
- Department of Pharmacological and Chemical Biology, Emory University, Atlanta, GA, United States
| | - Samarpan Majumder
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans (LSUHSC-NO), New Orleans, LA, United States
| | - Kristina Larter
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans (LSUHSC-NO), New Orleans, LA, United States
| | - Hanh Luu
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans (LSUHSC-NO), New Orleans, LA, United States
| | - Dorota Wyczechowska
- Department of Interdisciplinary Oncology, LSUHSC-NO, New Orleans, LA, United States
| | - Soroor Heidari
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans (LSUHSC-NO), New Orleans, LA, United States
| | - Keli Xu
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, MS, United States
| | - Sudarvili Shanthalingam
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, United States
| | | | - Yaguang Xi
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans (LSUHSC-NO), New Orleans, LA, United States
| | - Matthew Burow
- School of Medicine, Tulane University, New Orleans, LA, United States
| | | | - Luis Del Valle
- Department of Interdisciplinary Oncology, LSUHSC-NO, New Orleans, LA, United States
- Department of Pathology, Louisiana State University Health Sciences Center - New Orleans (LSUHSC-NO), New Orleans, LA, United States
| | - Chindo Hicks
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans (LSUHSC-NO), New Orleans, LA, United States
| | - Jovanny Zabaleta
- Department of Interdisciplinary Oncology, LSUHSC-NO, New Orleans, LA, United States
| | - Todd Golde
- Department of Pharmacological and Chemical Biology, Emory University, Atlanta, GA, United States
| | - Barbara Osborne
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, United States
| | - Lucio Miele
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans (LSUHSC-NO), New Orleans, LA, United States
| |
Collapse
|
7
|
Kumar R, Kumar R, Goel H, Kumar S, Ningombam SS, Haider I, Agrawal U, Deo S, Gogia A, Batra A, Sharma A, Mathur S, Ranjan A, Chopra A, Hussain S, Tanwar P. Whole exome sequencing identifies novel variants of PIK3CA and validation of hotspot mutation by droplet digital PCR in breast cancer among Indian population. Cancer Cell Int 2023; 23:236. [PMID: 37821962 PMCID: PMC10568783 DOI: 10.1186/s12935-023-03075-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 09/20/2023] [Indexed: 10/13/2023] Open
Abstract
BACKGROUND Breast cancer (BC) is the most common malignancy with very high incidence and relatively high mortality in women. The PIK3CA gene plays a pivotal role in the pathogenicity of breast cancer. Despite this, the mutational status of all exons except exons 9 and 20 still remains unknown. METHODS This study uses the whole exome sequencing (WES) based approach to identify somatic PIK3CA mutations in Indian BC cohorts. The resultant hotspot mutations were validated by droplet digital PCR (ddPCR). Further, molecular dynamics (MD) simulation was applied to elucidate the conformational and functional effects of hotspot position on PIK3CA protein. RESULTS In our cohort, PIK3CA showed a 44.4% somatic mutation rate and was among the top mutated genes. The mutations of PIK3CA were confined in Exons 5, 9, 11, 18, and 20, whereas the maximum number of mutations lies within exons 9 and 20. A total of 9 variants were found in our study, of which 2 were novel mutations observed on exons 9 (p.H554L) and 11 (p.S629P). However, H1047R was the hotspot mutation at exon 20 (20%). In tumor tissues, there was a considerable difference between copy number of wild-type and H1047R mutant was detected by ddPCR. Significant structural and conformational changes were observed during MD simulation, induced due to point mutation at H1047R/L position. CONCLUSIONS The current study provides a comprehensive view of novel as well as reported single nucleotide variants (SNVs) in PIK3CA gene associated with Indian breast cancer cases. The mutation status of H1047R/L could serve as a prognostic value in terms of selecting targeted therapy in BC.
Collapse
Affiliation(s)
- Rahul Kumar
- Dr B. R. A.-Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Rakesh Kumar
- Dr B. R. A.-Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Harsh Goel
- Dr B. R. A.-Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Sonu Kumar
- Department of Gastroenterology & HNU, All India Institute of Medical Sciences, New Delhi, India
| | - Somorjit Singh Ningombam
- Dr B. R. A.-Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Imran Haider
- Dr B. R. A.-Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Usha Agrawal
- National Institute of Pathology, New Delhi, India
| | - Svs Deo
- Dr B. R. A.-Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Ajay Gogia
- Dr B. R. A.-Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Atul Batra
- Dr B. R. A.-Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Ashok Sharma
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Sandeep Mathur
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Amar Ranjan
- Dr B. R. A.-Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Anita Chopra
- Dr B. R. A.-Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Showket Hussain
- Division of Molecular Oncology, National Institute of Cancer Prevention and Research, Noida, India
| | - Pranay Tanwar
- Dr B. R. A.-Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
8
|
Githaka JM, Pirayeshfard L, Goping IS. Cancer invasion and metastasis: Insights from murine pubertal mammary gland morphogenesis. Biochim Biophys Acta Gen Subj 2023; 1867:130375. [PMID: 37150225 DOI: 10.1016/j.bbagen.2023.130375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 04/20/2023] [Accepted: 05/02/2023] [Indexed: 05/09/2023]
Abstract
Cancer invasion and metastasis accounts for the majority of cancer related mortality. A better understanding of the players that drive the aberrant invasion and migration of tumors cells will provide critical targets to inhibit metastasis. Postnatal pubertal mammary gland morphogenesis is characterized by highly proliferative, invasive, and migratory normal epithelial cells. Identifying the molecular regulators of pubertal gland development is a promising strategy since tumorigenesis and metastasis is postulated to be a consequence of aberrant reactivation of developmental stages. In this review, we summarize the pubertal morphogenesis regulators that are involved in cancer metastasis and revisit pubertal mammary gland transcriptome profiling to uncover both known and unknown metastasis genes. Our updated list of pubertal morphogenesis regulators shows that most are implicated in invasion and metastasis. This review highlights molecular linkages between development and metastasis and provides a guide for exploring novel metastatic drivers.
Collapse
Affiliation(s)
- John Maringa Githaka
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada.
| | - Leila Pirayeshfard
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Ing Swie Goping
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada; Department of Oncology, University of Alberta, Edmonton, AB T6G 2H7, Canada.
| |
Collapse
|
9
|
Liang D, Gao Q, Meng Z, Li W, Song J, Xue K. Glycosylation in breast cancer progression and mammary development: Molecular connections and malignant transformations. Life Sci 2023; 326:121781. [PMID: 37207809 DOI: 10.1016/j.lfs.2023.121781] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 04/13/2023] [Accepted: 05/12/2023] [Indexed: 05/21/2023]
Abstract
INTRODUCTION The cellular behavior in normal mammary gland development and the progression of breast cancer is like the relationship between an object and its mirror image: they may appear similar, but their essence is completely different. Breast cancer can be considered as temporal and spatial aberrations of normal development in mammary gland. Glycans have been shown to regulate key pathophysiological steps during mammary development and breast cancer progression, and the glycoproteins that play a key role in both processes can affect the normal differentiation and development of mammary cells, and even cause malignant transformation or accelerate tumorigenesis due to differences in their type and level of glycosylation. KEY FINDINGS In this review, we summarize the roles of glycan alterations in essential cellular behaviors during breast cancer progression and mammary development, and also highlight the importance of key glycan-binding proteins such as epidermal growth factor receptor, transforming growth factor β receptors and other proteins, which are pivotal in the modulation of cellular signaling in mammary gland. Our review takes an overall view of the molecular interplay, signal transduction and cellular behaviors in mammary gland development and breast cancer progression from a glycobiological perspective. SIGNIFICANCE This review will give a better understanding of the similarities and differences in glycosylation between mammary gland development and breast cancer progression, laying the foundation for elucidating the key molecular mechanisms of glycobiology underlying the malignant transformation of mammary cells.
Collapse
Affiliation(s)
- Dongyang Liang
- College of Basic Medical Sciences, Dalian Medical University, Liaoning, China
| | - Qian Gao
- College of Basic Medical Sciences, Dalian Medical University, Liaoning, China
| | - Zixuan Meng
- College of Basic Medical Sciences, Dalian Medical University, Liaoning, China
| | - Wenzhe Li
- College of Basic Medical Sciences, Dalian Medical University, Liaoning, China
| | - Jiazhe Song
- College of Basic Medical Sciences, Dalian Medical University, Liaoning, China.
| | - Kai Xue
- College of Basic Medical Sciences, Dalian Medical University, Liaoning, China.
| |
Collapse
|
10
|
Monaco ML, Idris OA, Essani K. Triple-Negative Breast Cancer: Basic Biology and Immuno-Oncolytic Viruses. Cancers (Basel) 2023; 15:cancers15082393. [PMID: 37190321 DOI: 10.3390/cancers15082393] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/13/2023] [Accepted: 04/19/2023] [Indexed: 05/17/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is the most lethal subtype of breast cancer. TNBC diagnoses account for approximately one-fifth of all breast cancer cases globally. The lack of receptors for estrogen, progesterone, and human epidermal growth factor 2 (HER-2, CD340) results in a lack of available molecular-based therapeutics. This increases the difficulty of treatment and leaves more traditional as well as toxic therapies as the only available standards of care in many cases. Recurrence is an additional serious problem, contributing substantially to its higher mortality rate as compared to other breast cancers. Tumor heterogeneity also poses a large obstacle to treatment approaches. No driver of tumor development has been identified for TNBC, and large variations in mutational burden between tumors have been described previously. Here, we describe the biology of six different subtypes of TNBC, based on differential gene expression. Subtype differences can have a large impact on metastatic potential and resistance to treatment. Emerging antibody-based therapeutics, such as immune checkpoint inhibitors, have available targets for small subsets of TNBC patients, leading to partial responses and relatively low overall efficacy. Immuno-oncolytic viruses (OVs) have recently become significant in the pursuit of effective treatments for TNBC. OVs generally share the ability to ignore the heterogeneous nature of TNBC cells and allow infection throughout a treated tumor. Recent genetic engineering has allowed for the enhancement of efficacy against certain tumor types while avoiding the most common side effects in non-cancerous tissues. In this review, TNBC is described in order to address the challenges it presents to potential treatments. The OVs currently described preclinically and in various stages of clinical trials are also summarized, as are their strategies to enhance therapeutic potential.
Collapse
Affiliation(s)
- Michael L Monaco
- Laboratory of Virology, Department of Biological Sciences, Western Michigan University, Kalamazoo, MI 49008, USA
| | - Omer A Idris
- Laboratory of Virology, Department of Biological Sciences, Western Michigan University, Kalamazoo, MI 49008, USA
| | - Karim Essani
- Laboratory of Virology, Department of Biological Sciences, Western Michigan University, Kalamazoo, MI 49008, USA
| |
Collapse
|
11
|
Zhao X, Fu L, Zhai L, Yang X, Gao R. MiR-526b targets lncRNA SLC16A1-AS1 to suppress cell proliferation in triple-negative breast cancer. J Biochem Mol Toxicol 2023; 37:e23247. [PMID: 36654514 DOI: 10.1002/jbt.23247] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 07/31/2022] [Accepted: 10/12/2022] [Indexed: 01/20/2023]
Abstract
The present study investigated the potential interaction between miR-526b and lncRNA SLC16A1-AS1 in triple-negative breast cancer (TNBC). Expression of miR-526b and SLC16A1-AS1 in TNBC tumor tissues and paired nontumor tissues from 60 TNBC patients was detected by real-time polymerase chain reaction (RT-qPCR). The interaction between miR-526b and SLC16A1-AS1 was evaluated with overexpression experiments, followed by RT-qPCR. The proliferation and migration of cells were detected with cell counting kit-8 assay and Transwell assay, respectively. Apoptosis of cells was assessed by cell apoptosis assay. The expression of apoptosis-related proteins was quantified by Western blot analysis. MiR-526b was predicted to bind with SLC16A1-AS1. Overexpression of miR-526b in TNBC cells decreased the expression levels of SLC16A1-AS1, while overexpression of SLC16A1-AS1 did not affect the expression of miR-526b. In TNBC tissues, miR-526b was downregulated in TNBC tissues, while SLC16A1-AS1 was upregulated in TNBC tissues compared to that in nontumor tissues. The expression of SLC16A1-AS1 and miR-526b were inversely correlated. In vitro experiments showed that overexpression of SLC16A1-AS1 promoted cell proliferation and invasion but suppressed cell apoptosis. MiR-526b played an opposite role and suppressed the function of SLC16A1-AS1. MiR-526b is downregulated in TNBC and it targets SLC16A1-AS1 to regulate proliferation, apoptosis, and invasion of TNBC cells.
Collapse
Affiliation(s)
- Xingjuan Zhao
- Department of Mammary Gland, Shanxi People's Hospital, Taiyuan, Shanxi, China
| | - Lei Fu
- Department of Surgery, Shanxi Provincial General Team Hospital of the Chinese People's Armed Police Force, Taiyuan, Shanxi, China
| | - Liqin Zhai
- Department of Pathology, Shanxi People's Hospital, Shanxi, Taiyuan, China
| | - Xuan Yang
- Department of Mammary Gland, Shanxi People's Hospital, Taiyuan, Shanxi, China
| | - Runfang Gao
- Department of Mammary Gland, Shanxi People's Hospital, Taiyuan, Shanxi, China
| |
Collapse
|
12
|
Kaiser A, Eiselt G, Bechler J, Huber O, Schmidt M. WNT3a Signaling Inhibits Aromatase Expression in Breast Adipose Fibroblasts-A Possible Mechanism Supporting the Loss of Estrogen Responsiveness of Triple-Negative Breast Cancers. Int J Mol Sci 2023; 24:ijms24054654. [PMID: 36902090 PMCID: PMC10003471 DOI: 10.3390/ijms24054654] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 02/23/2023] [Accepted: 02/24/2023] [Indexed: 03/04/2023] Open
Abstract
Estrogen-dependent breast cancers rely on a constant supply of estrogens and expression of estrogen receptors. Local biosynthesis, by aromatase in breast adipose fibroblasts (BAFs), is their most important source for estrogens. Triple-negative breast cancers (TNBC) rely on other growth-promoting signals, including those from the Wnt pathway. In this study, we explored the hypothesis that Wnt signaling alters the proliferation of BAFs, and is involved in regulation of aromatase expression in BAFs. Conditioned medium (CM) from TNBC cells and WNT3a consistently increased BAF growth, and reduced aromatase activity up to 90%, by suppression of the aromatase promoter I.3/II region. Database searches identified three putative Wnt-responsive elements (WREs) in the aromatase promoter I.3/II. In luciferase reporter gene assays, promoter I.3/II activity was inhibited by overexpression of full-length T-cell factor (TCF)-4 in 3T3-L1 preadipocytes, which served as a model for BAFs. Full-length lymphoid enhancer-binding factor (LEF)-1 increased the transcriptional activity. However, TCF-4 binding to WRE1 in the aromatase promoter, was lost after WNT3a stimulation in immunoprecipitation-based in vitro DNA-binding assays, and in chromatin immunoprecipitation (ChIP). In vitro DNA-binding assays, ChIP, and Western blotting revealed a WNT3a-dependent switch of nuclear LEF-1 isoforms towards a truncated variant, whereas β-catenin levels remained unchanged. This LEF-1 variant revealed dominant negative properties, and most likely recruited enzymes involved in heterochromatin formation. In addition, WNT3a induced the replacement of TCF-4 by the truncated LEF-1 variant, on WRE1 of the aromatase promoter I.3/II. The mechanism described here may be responsible for the loss of aromatase expression predominantly associated with TNBC. Tumors with (strong) expression of Wnt ligands actively suppress aromatase expression in BAFs. Consequently a reduced estrogen supply could favor the growth of estrogen-independent tumor cells, which consequently would make estrogen receptors dispensable. In summary, canonical Wnt signaling within (cancerous) breast tissue may be a major factor controlling local estrogen synthesis and action.
Collapse
Affiliation(s)
- Alexander Kaiser
- Institute for Biochemistry II, Jena University Hospital, Friedrich Schiller University, 07743 Jena, Germany
| | - Gabriele Eiselt
- Institute for Biochemistry II, Jena University Hospital, Friedrich Schiller University, 07743 Jena, Germany
| | - Joachim Bechler
- Department of Gynecology and Obstetrics, Robert-Koch-Hospital, 99510 Apolda, Germany
| | - Otmar Huber
- Institute for Biochemistry II, Jena University Hospital, Friedrich Schiller University, 07743 Jena, Germany
| | - Martin Schmidt
- Institute for Biochemistry II, Jena University Hospital, Friedrich Schiller University, 07743 Jena, Germany
- Correspondence: ; Tel.: +49-3641-939-6420
| |
Collapse
|
13
|
Yamashita N, Kufe D. Addiction of Cancer Stem Cells to MUC1-C in Triple-Negative Breast Cancer Progression. Int J Mol Sci 2022; 23:8219. [PMID: 35897789 PMCID: PMC9331006 DOI: 10.3390/ijms23158219] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 07/20/2022] [Accepted: 07/21/2022] [Indexed: 02/01/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive malignancy with limited treatment options. TNBC progression is associated with expansion of cancer stem cells (CSCs). Few insights are available regarding druggable targets that drive the TNBC CSC state. This review summarizes the literature on TNBC CSCs and the compelling evidence that they are addicted to the MUC1-C transmembrane protein. In normal epithelia, MUC1-C is activated by loss of homeostasis and induces reversible wound-healing responses of inflammation and repair. However, in settings of chronic inflammation, MUC1-C promotes carcinogenesis. MUC1-C induces EMT, epigenetic reprogramming and chromatin remodeling in TNBC CSCs, which are dependent on MUC1-C for self-renewal and tumorigenicity. MUC1-C-induced lineage plasticity in TNBC CSCs confers DNA damage resistance and immune evasion by chronic activation of inflammatory pathways and global changes in chromatin architecture. Of therapeutic significance, an antibody generated against the MUC1-C extracellular domain has been advanced in a clinical trial of anti-MUC1-C CAR T cells and in IND-enabling studies for development as an antibody-drug conjugate (ADC). Agents targeting the MUC1-C cytoplasmic domain have also entered the clinic and are undergoing further development as candidates for advancing TNBC treatment. Eliminating TNBC CSCs will be necessary for curing this recalcitrant cancer and MUC1-C represents a promising druggable target for achieving that goal.
Collapse
Affiliation(s)
- Nami Yamashita
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Donald Kufe
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
14
|
Pincha N, Marangoni P, Haque A, Klein OD. Parallels in signaling between development and regeneration in ectodermal organs. Curr Top Dev Biol 2022; 149:373-419. [PMID: 35606061 PMCID: PMC10049776 DOI: 10.1016/bs.ctdb.2022.02.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Ectodermal organs originate from the outermost germ layer of the developing embryo and include the skin, hair, tooth, nails, and exocrine glands. These organs develop through tightly regulated, sequential and reciprocal epithelial-mesenchymal crosstalk, and they eventually assume various morphologies and functions while retaining the ability to regenerate. As with many other tissues in the body, the development and morphogenesis of these organs are regulated by a set of common signaling pathways, such as Shh, Wnt, Bmp, Notch, Tgf-β, and Eda. However, subtle differences in the temporal activation, the multiple possible combinations of ligand-receptor activation, the various cofactors, as well as the underlying epigenetic modulation determine how each organ develops into its adult form. Although each organ has been studied separately in considerable detail, the mechanisms underlying the parallels and differences in signaling that regulate their development have rarely been investigated. First, we will use the tooth, the hair follicle, and the mammary gland as representative ectodermal organs to explore how the development of signaling centers and establishment of stem cell populations influence overall growth and morphogenesis. Then we will compare how some of the major signaling pathways (Shh, Wnt, Notch and Yap/Taz) differentially regulate developmental events. Finally, we will discuss how signaling regulates regenerative processes in all three.
Collapse
Affiliation(s)
- Neha Pincha
- Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, CA, United States
| | - Pauline Marangoni
- Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, CA, United States
| | - Ameera Haque
- Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, CA, United States
| | - Ophir D Klein
- Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, CA, United States; Department of Pediatrics and Institute for Human Genetics, University of California, San Francisco, CA, United States.
| |
Collapse
|
15
|
Walker OL, Dahn ML, Power Coombs MR, Marcato P. The Prostaglandin E2 Pathway and Breast Cancer Stem Cells: Evidence of Increased Signaling and Potential Targeting. Front Oncol 2022; 11:791696. [PMID: 35127497 PMCID: PMC8807694 DOI: 10.3389/fonc.2021.791696] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 12/27/2021] [Indexed: 12/24/2022] Open
Abstract
Culprits of cancer development, metastasis, and drug resistance, cancer stem cells (CSCs) are characterized by specific markers, active developmental signaling pathways, metabolic plasticity, increased motility, invasiveness, and epithelial-mesenchymal transition. In breast cancer, these cells are often more prominent in aggressive disease, are amplified in drug-resistant tumors, and contribute to recurrence. For breast cancer, two distinct CSC populations exist and are typically defined by CD44+/CD24- cell surface marker expression or increased aldehyde dehydrogenase (ALDH) activity. These CSC populations share many of the same properties but also exhibit signaling pathways that are more active in CD44+/CD24- or ALDH+ populations. Understanding these CSC populations and their shared or specific signaling pathways may lead to the development of novel therapeutic strategies that will improve breast cancer patient outcomes. Herein, we review the current evidence and assess published patient tumor datasets of sorted breast CSC populations for evidence of heightened prostaglandin E2 (PGE2) signaling and activity in these breast CSC populations. PGE2 is a biologically active lipid mediator and in cancer PGE2 promotes tumor progression and poor patient prognosis. Overall, the data suggests that PGE2 signaling is important in propagating breast CSCs by enhancing inherent tumor-initiating capacities. Development of anti-PGE2 signaling therapeutics may be beneficial in inhibiting tumor growth and limiting breast CSC populations.
Collapse
Affiliation(s)
| | | | - Melanie R. Power Coombs
- Pathology, Dalhousie University, Halifax, NS, Canada
- Biology, Acadia University, Wolfville, NS, Canada
| | - Paola Marcato
- Pathology, Dalhousie University, Halifax, NS, Canada
- Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
- *Correspondence: Paola Marcato,
| |
Collapse
|
16
|
Zheng Y, Zhang J, Huang W, Zhong LLD, Wang N, Wang S, Yang B, Wang X, Pan B, Situ H, Lin Y, Liu X, Shi Y, Wang Z. Sini San Inhibits Chronic Psychological Stress-Induced Breast Cancer Stemness by Suppressing Cortisol-Mediated GRP78 Activation. Front Pharmacol 2021; 12:714163. [PMID: 34912211 PMCID: PMC8667778 DOI: 10.3389/fphar.2021.714163] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 11/05/2021] [Indexed: 11/13/2022] Open
Abstract
Chronic psychological stress is closely correlated with breast cancer growth and metastasis. Sini San (SNS) formula is a classical prescription for relieving depression-related symptoms in traditional Chinese medicine (TCM). Current researches have suggested that chronic psychological stress is closely correlated with cancer stem cells (CSCs) and endoplasmic reticulum (ER) stress. This study aimed to investigate the effects of chronic psychological stress on ER stress-mediated breast cancer stemness and the therapeutic implication of SNS. Chronic psychological stress promoted lung metastasis in 4T1 breast tumor-bearing mice and increased the stem cell-like populations and stemness-related gene expression. Meanwhile, GRP78, a marker of ER stress, was significantly increased in the breast tumors and lung metastases under chronic psychological stress. As a biochemical hallmark of chronic psychological stress, cortisol dramatically enhanced the stem cell-like populations and mammospheres formation by activating GRP78 transcriptionally. However, GRP78 inhibitors or shRNA attenuated the stemness enhancement mediated by cortisol. Similarly, SNS inhibited chronic psychological stress-induced lung metastasis and stemness of breast cancer cells, as well as reversed cortisol-induced stem cell-like populations and mammospheres formation by attenuating GRP78 expression. Co-localization and co-immunoprecipitation experiments showed that SNS interrupted the interaction between GRP78 and LRP5 on the cell surface, thus inhibiting the Wnt/β-catenin signaling of breast CSCs. Altogether, this study not only uncovers the biological influence and molecular mechanism of chronic psychological stress on breast CSCs but also highlights SNS as a promising strategy for relieving GRP78-induced breast cancer stemness via inhibiting GRP78 activation.
Collapse
Affiliation(s)
- Yifeng Zheng
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China.,State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Juping Zhang
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wanqing Huang
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Linda L D Zhong
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China.,School of Chinese Medicine, Hong Kong Baptist University, Kowloon, China
| | - Neng Wang
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China.,Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China.,The Research Center for Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shengqi Wang
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China.,Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China.,Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Bowen Yang
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xuan Wang
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Bo Pan
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Honglin Situ
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yi Lin
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaoyan Liu
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yafei Shi
- The Research Center for Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhiyu Wang
- The Research Center of Integrative Cancer Medicine, Discipline of Integrated Chinese and Western Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China.,State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China.,The Research Center for Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.,Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| |
Collapse
|
17
|
He L, Wick N, Germans SK, Peng Y. The Role of Breast Cancer Stem Cells in Chemoresistance and Metastasis in Triple-Negative Breast Cancer. Cancers (Basel) 2021; 13:cancers13246209. [PMID: 34944829 PMCID: PMC8699562 DOI: 10.3390/cancers13246209] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 12/05/2021] [Accepted: 12/08/2021] [Indexed: 02/05/2023] Open
Abstract
Triple negative breast cancer (TNBC) remains an aggressive disease due to the lack of targeted therapies and low rate of response to chemotherapy that is currently the main treatment modality for TNBC. Breast cancer stem cells (BCSCs) are a small subpopulation of breast tumors and recognized as drivers of tumorigenesis. TNBC tumors are characterized as being enriched for BCSCs. Studies have demonstrated the role of BCSCs as the source of metastatic disease and chemoresistance in TNBC. Multiple targets against BCSCs are now under investigation, with the considerations of either selectively targeting BCSCs or co-targeting BCSCs and non-BCSCs (majority of tumor cells). This review article provides a comprehensive overview of recent advances in the role of BCSCs in TNBC and the identification of cancer stem cell biomarkers, paving the way for the development of new targeted therapies. The review also highlights the resultant discovery of cancer stem cell targets in TNBC and the ongoing clinical trials treating chemoresistant breast cancer. We aim to provide insights into better understanding the mutational landscape of BCSCs and exploring potential molecular signaling pathways targeting BCSCs to overcome chemoresistance and prevent metastasis in TNBC, ultimately to improve the overall survival of patients with this devastating disease.
Collapse
Affiliation(s)
- Lin He
- Department of Pathology, University of Texas Southwestern Medical Center, 6201 Harry Hines Blvd, Dallas, TX 75235, USA; (L.H.); (N.W.); (S.K.G.)
| | - Neda Wick
- Department of Pathology, University of Texas Southwestern Medical Center, 6201 Harry Hines Blvd, Dallas, TX 75235, USA; (L.H.); (N.W.); (S.K.G.)
| | - Sharon Koorse Germans
- Department of Pathology, University of Texas Southwestern Medical Center, 6201 Harry Hines Blvd, Dallas, TX 75235, USA; (L.H.); (N.W.); (S.K.G.)
| | - Yan Peng
- Department of Pathology, University of Texas Southwestern Medical Center, 6201 Harry Hines Blvd, Dallas, TX 75235, USA; (L.H.); (N.W.); (S.K.G.)
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75235, USA
- Correspondence:
| |
Collapse
|
18
|
Luteolin Inhibits Breast Cancer Stemness and Enhances Chemosensitivity through the Nrf2-Mediated Pathway. Molecules 2021; 26:molecules26216452. [PMID: 34770867 PMCID: PMC8587415 DOI: 10.3390/molecules26216452] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/15/2021] [Accepted: 10/20/2021] [Indexed: 01/04/2023] Open
Abstract
Cancer stem cells (CSCs) are subpopulations of tumor masses with unique abilities in self-renewal, stemness maintenance, drug resistance, and the promotion of cancer recurrence. Recent studies have suggested that breast CSCs play essential roles in chemoresistance. Therefore, new agents that selectively target such cells are urgently required. Reactive oxygen species (ROS)-producing enzymes are the reason for an elevated tumor oxidant status. The nuclear factor erythroid 2-related factor 2 (Nrf2) is a transcriptional factor, which upon detecting cellular oxidative stress, binds to the promoter region of antioxidant genes. By triggering a cytoprotective response, Nrf2 maintains cellular redox status. Cripto-1 participates in the self-renewal of CSCs. Herein, luteolin, a flavonoid found in Taraxacum officinale extract, was determined to inhibit the expressions of stemness-related transcriptional factors, the ATP-binding cassette transporter G2 (ABCG2), CD44, aldehyde dehydrogenase 1 activity as well as the sphere formation properties of breast CSCs. Furthermore, luteolin suppressed the protein expressions of Nrf2, heme oxygenase 1 (HO-1), and Cripto-1 which have been determined to contribute critically to CSC features. The combination of luteolin and the chemotherapeutic drug, Taxol, resulted in enhanced cytotoxicity to breast cancer cells. These findings suggest that luteolin treatment significantly attenuated the hallmarks of breast cancer stemness by downregulating Nrf2-mediated expressions. Luteolin constitutes a potential agent for use in cancer stemness-targeted breast cancer treatments.
Collapse
|
19
|
Casey T, Suarez-Trujillo A, Cummings S, Huff K, Crodian J, Bhide K, Aduwari C, Teeple K, Shamay A, Mabjeesh SJ, San Miguel P, Thimmapuram J, Plaut K. Core circadian clock transcription factor BMAL1 regulates mammary epithelial cell growth, differentiation, and milk component synthesis. PLoS One 2021; 16:e0248199. [PMID: 34415905 PMCID: PMC8378744 DOI: 10.1371/journal.pone.0248199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 08/06/2021] [Indexed: 11/18/2022] Open
Abstract
The role the mammary epithelial circadian clock plays in gland development and lactation is unknown. We hypothesized that mammary epithelial clocks function to regulate mammogenesis and lactogenesis, and propose the core clock transcription factor BMAL1:CLOCK regulates genes that control mammary epithelial development and milk synthesis. Our objective was to identify transcriptional targets of BMAL1 in undifferentiated (UNDIFF) and lactogen differentiated (DIFF) mammary epithelial cells (HC11) using ChIP-seq. Ensembl gene IDs with the nearest transcriptional start site to ChIP-seq peaks were explored as potential targets, and represented 846 protein coding genes common to UNDIFF and DIFF cells and 2773 unique to DIFF samples. Genes with overlapping peaks between samples (1343) enriched cell-cell adhesion, membrane transporters and lipid metabolism categories. To functionally verify targets, an HC11 line with Bmal1 gene knocked out (BMAL1-KO) using CRISPR-CAS was created. BMAL1-KO cultures had lower cell densities over an eight-day growth curve, which was associated with increased (p<0.05) levels of reactive oxygen species and lower expression of superoxide dismutase 3 (Sod3). RT-qPCR analysis also found lower expression of the putative targets, prolactin receptor (Prlr), Ppara, and beta-casein (Csn2). Findings support our hypothesis and highlight potential importance of clock in mammary development and substrate transport.
Collapse
Affiliation(s)
- Theresa Casey
- Department of Animal Science, Purdue University, West Lafayette, IN, United States of America
| | - Aridany Suarez-Trujillo
- Department of Animal Science, Purdue University, West Lafayette, IN, United States of America
| | - Shelby Cummings
- Department of Animal Science, Purdue University, West Lafayette, IN, United States of America
| | - Katelyn Huff
- Department of Animal Science, Purdue University, West Lafayette, IN, United States of America
| | - Jennifer Crodian
- Department of Animal Science, Purdue University, West Lafayette, IN, United States of America
| | - Ketaki Bhide
- Bioinformatics Core, Purdue University, West Lafayette, IN, United States of America
| | - Clare Aduwari
- Department of Animal Science, Purdue University, West Lafayette, IN, United States of America
| | - Kelsey Teeple
- Department of Animal Science, Purdue University, West Lafayette, IN, United States of America
| | - Avi Shamay
- Animal Science Institute, Agriculture Research Origination, The Volcani Center, Rishon Letsiyon, Israel
| | - Sameer J. Mabjeesh
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food, and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Phillip San Miguel
- Genomics Core, Purdue University, West Lafayette, IN, United States of America
| | - Jyothi Thimmapuram
- Bioinformatics Core, Purdue University, West Lafayette, IN, United States of America
| | - Karen Plaut
- Department of Animal Science, Purdue University, West Lafayette, IN, United States of America
| |
Collapse
|
20
|
Ling B, Liao X, Tang Q, Ye G, Bin X, Wang J, Pang Y, Qi G. MicroRNA-106b-5p inhibits growth and progression of lung adenocarcinoma cells by downregulating IGSF10. Aging (Albany NY) 2021; 13:18740-18756. [PMID: 34351868 PMCID: PMC8351668 DOI: 10.18632/aging.203318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 05/18/2021] [Indexed: 12/04/2022]
Abstract
In this study, we investigated the mechanistic role and prognostic significance of IGSF10 in lung adenocarcinoma. Oncomine database analysis showed that IGSF10 expression was significantly reduced in most cancer types, including lung adenocarcinoma (LUAD). In the TCGA-LUAD dataset, IGSF10 expression correlated positively with proportions of tumor-infiltrated B cells, CD4+ T cells, CD8+ T cells, neutrophils, macrophages, and dendritic cells. Kaplan-Meier survival analysis showed that overall survival of patients with low IGSF10 expression was significantly shorter than those with high IGSF10 expression. MiRWalk2.0 database analysis and dual luciferase reporter assays confirmed that miR-106b-5p suppressed IGSF10 expression by binding to its 3’UTR. MiR-106b-5p levels inversely correlated with IGSF10 expression in the TCGA-LUAD dataset. Moreover, inhibition of miR-106b-5p significantly decreased in vitro proliferation, migration, and invasion by LUAD cells, whereas miR-106b-5p overexpression reversed those effects. These results demonstrate that IGSF10 is an independent prognostic factor for LUAD. Furthermore, miR-106b-5p suppressed IGSF10 expression in LUAD tissues by binding to its 3’UTR, which makes IGSF10 and miR-106b-5p potential prognostic biomarkers and therapeutic targets in LUAD patients.
Collapse
Affiliation(s)
- Bo Ling
- College of Pharmacy, Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Xianjiu Liao
- College of Pharmacy, Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Qiang Tang
- Department of Burn and Plastic Surgery and Wound Repair, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Guangbin Ye
- College of Basic Medical Sciences, Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China.,Medical College of Guangxi University, Nanning 530004, Guangxi, China
| | - Xiaoyun Bin
- College of Basic Medical Sciences, Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Jianchu Wang
- Department of Hepatobiliary Surgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Yaqin Pang
- College of Medical Laboratory, Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Guangzi Qi
- College of Public Health and Management, Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| |
Collapse
|
21
|
Githaka JM, Tripathi N, Kirschenman R, Patel N, Pandya V, Kramer DA, Montpetit R, Zhu LF, Sonenberg N, Fahlman RP, Danial NN, Underhill DA, Goping IS. BAD regulates mammary gland morphogenesis by 4E-BP1-mediated control of localized translation in mouse and human models. Nat Commun 2021; 12:2939. [PMID: 34011960 PMCID: PMC8134504 DOI: 10.1038/s41467-021-23269-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 04/20/2021] [Indexed: 02/03/2023] Open
Abstract
Elucidation of non-canonical protein functions can identify novel tissue homeostasis pathways. Herein, we describe a role for the Bcl-2 family member BAD in postnatal mammary gland morphogenesis. In Bad3SA knock-in mice, where BAD cannot undergo phosphorylation at 3 key serine residues, pubertal gland development is delayed due to aberrant tubulogenesis of the ductal epithelium. Proteomic and RPPA analyses identify that BAD regulates focal adhesions and the mRNA translation repressor, 4E-BP1. These results suggest that BAD modulates localized translation that drives focal adhesion maturation and cell motility. Consistent with this, cells within Bad3SA organoids contain unstable protrusions with decreased compartmentalized mRNA translation and focal adhesions, and exhibit reduced cell migration and tubulogenesis. Critically, protrusion stability is rescued by 4E-BP1 depletion. Together our results confirm an unexpected role of BAD in controlling localized translation and cell migration during mammary gland development.
Collapse
Affiliation(s)
- John Maringa Githaka
- grid.17089.37Department of Biochemistry, University of Alberta, Edmonton, AB Canada
| | - Namita Tripathi
- grid.17089.37Department of Biochemistry, University of Alberta, Edmonton, AB Canada
| | - Raven Kirschenman
- grid.17089.37Department of Biochemistry, University of Alberta, Edmonton, AB Canada
| | - Namrata Patel
- grid.17089.37Department of Biochemistry, University of Alberta, Edmonton, AB Canada
| | - Vrajesh Pandya
- grid.17089.37Department of Biochemistry, University of Alberta, Edmonton, AB Canada
| | - David A. Kramer
- grid.17089.37Department of Biochemistry, University of Alberta, Edmonton, AB Canada
| | - Rachel Montpetit
- grid.17089.37Department of Biochemistry, University of Alberta, Edmonton, AB Canada
| | - Lin Fu Zhu
- grid.17089.37Department of Surgery, University of Alberta, Edmonton, AB Canada
| | - Nahum Sonenberg
- grid.14709.3b0000 0004 1936 8649Department of Biochemistry, McGill University, Montreal, QC Canada
| | - Richard P. Fahlman
- grid.17089.37Department of Biochemistry, University of Alberta, Edmonton, AB Canada
| | - Nika N. Danial
- grid.65499.370000 0001 2106 9910Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA USA
| | - D. Alan Underhill
- grid.17089.37Department of Oncology, University of Alberta, Edmonton, AB Canada
| | - Ing Swie Goping
- grid.17089.37Department of Biochemistry, University of Alberta, Edmonton, AB Canada ,grid.17089.37Department of Oncology, University of Alberta, Edmonton, AB Canada
| |
Collapse
|
22
|
GATA3 somatic mutations are associated with clinicopathological features and expression profile in TCGA breast cancer patients. Sci Rep 2021; 11:1679. [PMID: 33462316 PMCID: PMC7814117 DOI: 10.1038/s41598-020-80680-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 12/22/2020] [Indexed: 12/11/2022] Open
Abstract
The effect of somatic mutations and the gene expression profiles on the prognosis is well documented in cancer research. This study was conducted to evaluate the association of GATA3 somatic mutations with tumor features, survival, and expression profiles in breast cancer. Clinicopathological information was compared between TCGA-BRCA patients with GATA3-mutant and non-mutant tumors in all patients as well as in ER-positive subgroup. Cox-regression method was used to evaluate the association of the GATA3 mutation status with overall survival time. Differential gene expression, functional annotation, and protein-protein interaction analyses were performed using edgeR, Metascape, DAVID, STRING and CytoNCA. GATA3-mutant and non-mutant samples had significantly different clinicopathological features (p < 0.05). While GATA3 mutation status was not associated with the overall survival in the entire cohort (padj = 0.52), the GATA3-wild type ER-positive cases had a better prognosis than mutant ones (padj = 0.04). GATA3 expression was higher in tumors than normal tissues. Several pathways were different between mutant and non-mutant groups (p < 0.05). Interleukin-6 was found as the highest scored gene in both comparisons (normal vs. mutant and normal vs. non-mutant groups) in the entire patient and in the ER-positive subgroup, suggesting the association of IL6 with breast tumorigenesis. These findings suggest that GATA3 mutations can be associated with several tumor characteristics and influence the pattern of gene expression. However, GATA3 mutation status seems to be a prognostic factor for the disease only in ER-positive patients.
Collapse
|
23
|
Kumar S, Nandi A, Singh S, Regulapati R, Li N, Tobias JW, Siebel CW, Blanco MA, Klein-Szanto AJ, Lengner C, Welm AL, Kang Y, Chakrabarti R. Dll1 + quiescent tumor stem cells drive chemoresistance in breast cancer through NF-κB survival pathway. Nat Commun 2021; 12:432. [PMID: 33462238 PMCID: PMC7813834 DOI: 10.1038/s41467-020-20664-5] [Citation(s) in RCA: 44] [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: 01/13/2020] [Accepted: 12/10/2020] [Indexed: 01/29/2023] Open
Abstract
Development of chemoresistance in breast cancer patients greatly increases mortality. Thus, understanding mechanisms underlying breast cancer resistance to chemotherapy is of paramount importance to overcome this clinical challenge. Although activated Notch receptors have been associated with chemoresistance in cancer, the specific Notch ligands and their molecular mechanisms leading to chemoresistance in breast cancer remain elusive. Using conditional knockout and reporter mouse models, we demonstrate that tumor cells expressing the Notch ligand Dll1 is important for tumor growth and metastasis and bear similarities to tumor-initiating cancer cells (TICs) in breast cancer. RNA-seq and ATAC-seq using reporter models and patient data demonstrated that NF-κB activation is downstream of Dll1 and is associated with a chemoresistant phenotype. Finally, pharmacological blocking of Dll1 or NF-κB pathway completely sensitizes Dll1+ tumors to chemotherapy, highlighting therapeutic avenues for chemotherapy resistant breast cancer patients in the near future.
Collapse
Affiliation(s)
- Sushil Kumar
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ajeya Nandi
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Snahlata Singh
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Rohan Regulapati
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ning Li
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - John W Tobias
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Christian W Siebel
- Department of Discovery Oncology, Genentech Inc., South San Francisco, CA, 94080, USA
| | - Mario Andres Blanco
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | | | - Christopher Lengner
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Alana L Welm
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, 84112, USA
| | - Yibin Kang
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA
| | - Rumela Chakrabarti
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
24
|
Silva AF, Silva ECCDA, Oliveira CN, Rocha KBF, Miguel MCC, Pereira JS. Oral and uterine leiomyomas exhibit high immunoexpression of Cripto-1 compared to normal myometrium. AN ACAD BRAS CIENC 2020; 92:e20200519. [PMID: 33331446 DOI: 10.1590/0001-3765202020200519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 09/21/2020] [Indexed: 11/22/2022] Open
Abstract
Leiomyomas are the most common benign tumors in women. Many of them are associated with significant morbidity. The present study aimed to analyze histomorphological and histochemical characteristics and immunoexpression of Cripto-1 in oral leiomyomas (OL), uterine leiomyomas (UL), and normal myometrium (NM). Sample was composed of ten OL, 11 UL and 11 NM. Histomorphological characteristics were analyzed at 100 and 400x magnifications with HE staining. The immunoexpression of Cripto-1 was analyzed in five high-power fields. Statistical analysis considered a significant difference when p<0.05. Six OL disclosed moderate/intense inflammatory infiltrate, while ten UL exhibited mild infiltrate (p=0.024). When analyzing all leiomyomas together, 20 exhibited hyalinization, whereas no NM exhibited this alteration (p<0.001). There was no statistical difference in the distribution of mast cells among the lesions. The median Cripto-1 was higher in UL (9.0), followed by OL and NM (4.0). Associations of the Cripto-1 expression between leiomyomas (separately and together) and NM were statistically significant (p<0.001). These results indicate that OL and UL exhibit similar histomorphological and histochemical characteristics, as well as differences to NM. The higher immunoexpression of Cripto-1 in leiomyomas compared to NM suggests that this protein may influence cell proliferation and tissue architecture of oral and uterine leiomyomas.
Collapse
Affiliation(s)
- AndrÉ F Silva
- Universidade Federal do Rio Grande do Norte, Departamento de Patologia, Rua General Gustavo Cordeiro de Farias, s/n, Petrópolis, 59012-570 Natal, RN, Brazil
| | - Emmily C C DA Silva
- Universidade Federal do Rio Grande do Norte, Departamento de Patologia, Rua General Gustavo Cordeiro de Farias, s/n, Petrópolis, 59012-570 Natal, RN, Brazil
| | - ClÁudia N Oliveira
- Universidade Federal do Rio Grande do Norte, Departamento de Patologia, Rua General Gustavo Cordeiro de Farias, s/n, Petrópolis, 59012-570 Natal, RN, Brazil
| | - Keyla B F Rocha
- Universidade Federal do Rio Grande do Norte, Departamento de Patologia, Rua General Gustavo Cordeiro de Farias, s/n, Petrópolis, 59012-570 Natal, RN, Brazil
| | - Marcia C C Miguel
- Programa de Pós-Graduação em Patologia Oral, Universidade Federal do Rio Grande do Norte, Rua Senador Salgado Filho, 1787, Lagoa Nova, 59056-000 Natal, RN, Brazil
| | - Joabe S Pereira
- Universidade Federal do Rio Grande do Norte, Departamento de Patologia, Rua General Gustavo Cordeiro de Farias, s/n, Petrópolis, 59012-570 Natal, RN, Brazil
| |
Collapse
|
25
|
Sriroopreddy R, Raghuraman P, Sreeshma J, Kamalesh D, Sudandiradoss C. Exploring the structural significance of molecular switch mechanism alias motif phosphorylation in Wnt/β-catenin and their crucial role in triple-negative breast cancer. Arch Biochem Biophys 2020; 698:108722. [PMID: 33321112 DOI: 10.1016/j.abb.2020.108722] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 11/20/2020] [Accepted: 12/07/2020] [Indexed: 11/25/2022]
Abstract
β-Catenin, a key transcriptional factor involved in the canonical Wnt signaling pathway, is regulated by a cascade of phosphorylations and plays a major role in the progression of triple-negative breast cancer (TNBC). However, the phosphorylation induced conformational changes in a β-Catenin is still poorly understood. Hence, we adopted a conventional molecular dynamics approach to study phosphorylations present in a sequence motif Ser 552 675 and Tyr670 of the β-Catenin domain and analyzed in terms of structural transitions, bond formation, and folding-misfolding conformations. Our results unveil the β-Catenin linear motif 549-555 (RRTSMGG) of armadillo repeats domain prefers order to disorder state. In contrast, helix C associated with 670-678 (YKKRLSVEL) motif prefers disorder to order upon phosphorylation of Ser 552 675 and Tyr670. In addition, the crucial secondary structural transition from α-helix to coil induced by phospho Ser552 and phospho Tyr670 of β-Catenin ARM domain connecting helix C modifies conformational diversity and binding affinities of the complex interaction in functional regulation significantly. Moreover, the post phosphorylation disrupted the hydrogen bond interactions (Ser552-Arg549, Arg550-Asp546 and Ser675-Lys672) and abolished the residual alliance with hydrophobic interactions (Tyr670-Leu674) that easily interrupt in secondary structure packing as well as folding conformations connecting ARM and helix C (R10, 12 & R1C) compared to unphosphorylation. Our integrated computational analysis may help in shedding light on understanding the induced folding and unfolding pattern due to motif phosphorylations. Overall, our results provide an atomistic structural description of the way phosphorylation facilitates conformational and dynamic changes in β-Catenin, a fundamental molecular switch mechanism in triple-negative breast cancer pathogenesis.
Collapse
Affiliation(s)
- Ramireddy Sriroopreddy
- Department of Biotechnology, School of Bioscience and Technology, Vellore Institute of Technology, Vellore, 632014, India
| | - P Raghuraman
- Department of Biotechnology, School of Bioscience and Technology, Vellore Institute of Technology, Vellore, 632014, India
| | - J Sreeshma
- Department of Biotechnology, School of Bioscience and Technology, Vellore Institute of Technology, Vellore, 632014, India
| | - D Kamalesh
- Department of Biotechnology, School of Bioscience and Technology, Vellore Institute of Technology, Vellore, 632014, India
| | - C Sudandiradoss
- Department of Biotechnology, School of Bioscience and Technology, Vellore Institute of Technology, Vellore, 632014, India.
| |
Collapse
|
26
|
Xu H, Yang X, Huang W, Ma Y, Ke H, Zou L, Yang Q, Jiao B. Single-cell profiling of long noncoding RNAs and their cell lineage commitment roles via RNA-DNA-DNA triplex formation in mammary epithelium. Stem Cells 2020; 38:1594-1611. [PMID: 32930441 DOI: 10.1002/stem.3274] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 07/21/2020] [Accepted: 08/03/2020] [Indexed: 06/11/2023]
Abstract
Long noncoding RNAs (lncRNAs), which are crucial for organ development, exhibit cell-specific expression. Thus, transcriptomic analysis based on total tissue (bulk-seq) cannot accurately reflect the expression pattern of lncRNAs. Here, we used high-throughput single-cell RNA-seq data to investigate the role of lncRNAs using the hierarchical model of mammary epithelium. With our comprehensive annotation of the mammary epithelium, lncRNAs showed much greater cell-lineage specific expression than coding genes. The lineage-specific lncRNAs were functionally correlated with lineage commitment through the coding genes via the cis- and trans-effects of lncRNAs. For the working mechanism, lncRNAs formed a triplex structure with the DNA helix to regulate downstream lineage-specific marker genes. We used lncRNA-Carmn as an example to validate the above findings. Carmn, which is specifically expressed in mammary gland stem cells (MaSCs) and basal cells, positively regulated the Wnt signaling ligand Wnt10a through formation of a lncRNA-DNA-DNA triplex, and thus controlled the stemness of MaSCs. Our study suggests that lncRNAs play essential roles in cell-lineage commitment and provides an approach to decipher lncRNA functions based on single-cell RNA-seq data.
Collapse
Affiliation(s)
- Haibo Xu
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, People's Republic of China
- Department of Urology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, People's Republic of China
- International Cancer Center, Shenzhen University School of Medicine, Shenzhen, People's Republic of China
| | - Xing Yang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, People's Republic of China
- Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, People's Republic of China
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, Yunnan, People's Republic of China
| | - Weiren Huang
- Department of Urology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, People's Republic of China
- International Cancer Center, Shenzhen University School of Medicine, Shenzhen, People's Republic of China
| | - Yujie Ma
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, People's Republic of China
| | - Hao Ke
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, People's Republic of China
| | - Li Zou
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, People's Republic of China
| | - Qin Yang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, People's Republic of China
| | - Baowei Jiao
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, People's Republic of China
- KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, People's Republic of China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, Yunnan, People's Republic of China
| |
Collapse
|
27
|
Retinoic acid induces differentiation in neuroblastoma via ROR1 by modulating retinoic acid response elements. Oncol Rep 2020; 44:1013-1024. [PMID: 32705280 PMCID: PMC7388440 DOI: 10.3892/or.2020.7681] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 06/16/2020] [Indexed: 12/14/2022] Open
Abstract
Neuroblastoma is the most common inheritable, solid neoplasm in children found under the age of 7 and accounts for approximately 7% of childhood cancers. A common treatment that has been prescribed for over a decade is retinoid therapy [using all-trans retinoic acid (RA)]. Treatment with this differentiating agent has been revealed to progress the cells from their stem-cell state to a mature neuronal state gaining classical neuronal characteristics, including the suppression of proliferation. However, the molecular mechanism underlying the action of RA treatment remains to be elucidated. In the present study, a novel mechanism of RA-induced differentiation via regulation of receptor tyrosine kinase-like orphan receptor 1 (ROR1) is reported. ROR1 is overexpressed in neuroblastoma but significantly downregulated in mature differentiated neurons. Hence, it was hypothesized that RA may modulate ROR1 leading to differentiation and termination of cancerous properties. Immunoblotting revealed that following RA treatment, ROR1 levels initially increased then sharply decreased by 96 h. This was paired with synaptophysin, a mature neuron marker, sharply increasing concurrently, providing evidence of differentiation by 96 h. Investigation of the ROR1 pathway confirmed ROR1-dependent downstream activation of the PI3K/AKT signaling axis, a growth pathway previously demonstrated to promote differentiation. Chromatin immunoprecipitation revealed an increase in RAR binding to the promoters of ROR1 and its endogenous ligand, Wnt5a. This research provided compelling evidence that RA is able to modulate the expression of ROR1 and Wnt5a to promote differentiation through the expression of synaptophysin. This data combined with the overarching data from the scientific community regarding proliferation and other proliferative factors in early-stage neurons provides a more in-depth model of the process of differentiation in neurons.
Collapse
|
28
|
Exploiting Cancer's Tactics to Make Cancer a Manageable Chronic Disease. Cancers (Basel) 2020; 12:cancers12061649. [PMID: 32580319 PMCID: PMC7352192 DOI: 10.3390/cancers12061649] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 06/17/2020] [Accepted: 06/19/2020] [Indexed: 12/26/2022] Open
Abstract
The history of modern oncology started around eighty years ago with the introduction of cytotoxic agents such as nitrogen mustard into the clinic, followed by multi-agent chemotherapy protocols. Early success in radiation therapy in Hodgkin lymphoma gave birth to the introduction of radiation therapy into different cancer treatment protocols. Along with better understanding of cancer biology, we developed drugs targeting cancer-related cellular and genetic aberrancies. Discovery of the crucial role of vasculature in maintenance, survival, and growth of a tumor opened the way to the development of anti-angiogenic agents. A better understanding of T-cell regulatory pathways advanced immunotherapy. Awareness of stem-like cancer cells and their role in cancer metastasis and local recurrence led to the development of drugs targeting them. At the same time, sequential and rapidly accelerating advances in imaging and surgical technology have markedly increased our ability to safely remove ≥90% of tumor cells. While we have advanced our ability to kill cells from multiple directions, we have still failed to stop most types of cancer from recurring. Here we analyze the tactics employed in cancer evolution; namely, chromosomal instability (CIN), intra-tumoral heterogeneity (ITH), and cancer-specific metabolism. These tactics govern the resistance to current cancer therapeutics. It is time to focus on maximally delaying the time to recurrence, with drugs that target these fundamental tactics of cancer evolution. Understanding the control of CIN and the optimal state of ITH as the most important tactics in cancer evolution could facilitate the development of improved cancer therapeutic strategies designed to transform cancer into a manageable chronic disease.
Collapse
|
29
|
Oliphant MUJ, Kong D, Zhou H, Lewis MT, Ford HL. Two Sides of the Same Coin: The Role of Developmental pathways and pluripotency factors in normal mammary stem cells and breast cancer metastasis. J Mammary Gland Biol Neoplasia 2020; 25:85-102. [PMID: 32323111 PMCID: PMC7395869 DOI: 10.1007/s10911-020-09449-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 03/25/2020] [Indexed: 02/06/2023] Open
Abstract
Breast cancer initiation and progression are often observed as the result of dysregulation of normal developmental processes and pathways. Studies focused on normal mammary stem/progenitor cell activity have led to an understanding of how breast cancer cells acquire stemness-associated properties including tumor initiation, survival and multi-lineage differentiation into heterogeneous tumors that become difficult to target therapeutically. Importantly, more recent investigations have provided valuable insight into how key developmental regulators can impact multiple phases of metastasis, where they are repurposed to not only promote metastatic phenotypes such as migration, invasion and EMT at the primary site, but also to regulate the survival, initiation and maintenance of metastatic lesions at secondary organs. Herein, we discuss findings that have led to a better understanding of how embryonic and pluripotency factors contribute not only to normal mammary development, but also to metastatic progression. We further examine the therapeutic potential of targeting these developmental pathways, and discuss how a better understanding of compensatory mechanisms, crosstalk between pathways, and novel experimental models could provide critical insight into how we might exploit embryonic and pluripotency regulators to inhibit tumor progression and metastasis.
Collapse
Affiliation(s)
- M U J Oliphant
- Integrated Physiology Program, University of Colorado Anschutz Medical Campus, RC1-North, P18-6115, 12800 East 19th Ave, Aurora, CO, 80045, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, RC1-North, P18-6115, 12800 East 19th Ave, Aurora, CO, 80045, USA
- Department of Cell Biology and Ludwig Center at Harvard, Harvard Medical School, 240 Longwood Avenue, Building C1, Room 513B, Boston, MA, 02115, USA
| | - Deguang Kong
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, RC1-North, P18-6115, 12800 East 19th Ave, Aurora, CO, 80045, USA
| | - Hengbo Zhou
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, RC1-North, P18-6115, 12800 East 19th Ave, Aurora, CO, 80045, USA
- Cancer Biology Program, University of Colorado Anschutz Medical Campus, RC1-North, P18-6115, 12800 East 19th Ave, Aurora, CO, 80045, USA
| | - M T Lewis
- Departments of Molecular and Cellular Biology and Radiology. Lester and Sue Smith Breast Center, Baylor College of Medicine. One Baylor Plaza BCM600, Room N1210, Houston, TX, 77030, USA
| | - H L Ford
- Integrated Physiology Program, University of Colorado Anschutz Medical Campus, RC1-North, P18-6115, 12800 East 19th Ave, Aurora, CO, 80045, USA.
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, RC1-North, P18-6115, 12800 East 19th Ave, Aurora, CO, 80045, USA.
- Cancer Biology Program, University of Colorado Anschutz Medical Campus, RC1-North, P18-6115, 12800 East 19th Ave, Aurora, CO, 80045, USA.
| |
Collapse
|
30
|
Antibody-drug conjugates targeting RON receptor tyrosine kinase as a novel strategy for treatment of triple-negative breast cancer. Drug Discov Today 2020; 25:1160-1173. [PMID: 32479905 DOI: 10.1016/j.drudis.2020.05.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 04/05/2020] [Accepted: 05/18/2020] [Indexed: 12/17/2022]
Abstract
Treatment of triple-negative breast cancer (TNBC) is a challenge to oncologists. Currently, the lack of effective therapy has fostered a major effort to discover new targets and therapeutics to combat this disease. The recepteur d'origine nantais (RON) receptor has been implicated in the pathogenesis of TNBC. Clinical studies have revealed that aberrant RON expression is crucial in regulating TNBC malignant phenotypes. Increased RON expression also has prognostic value for breast cancer progress. These features provide the rationale to target RON for TNBC treatment. In this review, we discuss the importance of RON in TNBC tumorigenesis and the development of anti-RON antibody-drug conjugates (ADCs) for clinical application. The findings from preclinical studies lay the foundation for clinical trials of this novel biotherapeutic for TNBC therapy.
Collapse
|
31
|
Yu K, Rohr J, Liu Y, Li M, Xu J, Wang K, Chai J, Zhao D, Liu Y, Ma J, Fan L, Wang Z, Guo S. Progress in triple negative breast carcinoma pathophysiology: Potential therapeutic targets. Pathol Res Pract 2020; 216:152874. [PMID: 32088086 DOI: 10.1016/j.prp.2020.152874] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 01/17/2020] [Accepted: 02/11/2020] [Indexed: 12/17/2022]
Abstract
Triple-negative breast carcinoma (TNBC) is a subtype of breast carcinoma defined by negativity for estrogen receptor (ER) or progesterone receptor (PR) by immunohistochemical analysis and negativity for human epidermal growth factor receptor (Her2) by immunohistochemistry or in situ hybridization. TNBC is clinically marked by its high aggressiveness, particularly poor outcomes including a low survival rate, and the lack of specific and effective treatments. Therefore, new potential targets for the treatment of TNBC must be identified. This review summarizes recent evidence supporting novel targets and possible therapeutic regimens in the treatment of TNBC.
Collapse
Affiliation(s)
- Kangjie Yu
- State Key Laboratory of Tumor Biology, Department of Pathology, Xi Jing Hospital, the Fourth Military Medical University, Xi'an, Shaan Xi Province,710032, China
| | - Joseph Rohr
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Yang Liu
- State Key Laboratory of Tumor Biology, Department of Pathology, Xi Jing Hospital, the Fourth Military Medical University, Xi'an, Shaan Xi Province,710032, China
| | - Mingyang Li
- State Key Laboratory of Tumor Biology, Department of Pathology, Xi Jing Hospital, the Fourth Military Medical University, Xi'an, Shaan Xi Province,710032, China
| | - Junpeng Xu
- State Key Laboratory of Tumor Biology, Department of Pathology, Xi Jing Hospital, the Fourth Military Medical University, Xi'an, Shaan Xi Province,710032, China
| | - Kaijing Wang
- State Key Laboratory of Tumor Biology, Department of Pathology, Xi Jing Hospital, the Fourth Military Medical University, Xi'an, Shaan Xi Province,710032, China
| | - Jia Chai
- State Key Laboratory of Tumor Biology, Department of Pathology, Xi Jing Hospital, the Fourth Military Medical University, Xi'an, Shaan Xi Province,710032, China
| | - Danhui Zhao
- State Key Laboratory of Tumor Biology, Department of Pathology, Xi Jing Hospital, the Fourth Military Medical University, Xi'an, Shaan Xi Province,710032, China
| | - Yixiong Liu
- State Key Laboratory of Tumor Biology, Department of Pathology, Xi Jing Hospital, the Fourth Military Medical University, Xi'an, Shaan Xi Province,710032, China
| | - Jing Ma
- State Key Laboratory of Tumor Biology, Department of Pathology, Xi Jing Hospital, the Fourth Military Medical University, Xi'an, Shaan Xi Province,710032, China
| | - Linni Fan
- State Key Laboratory of Tumor Biology, Department of Pathology, Xi Jing Hospital, the Fourth Military Medical University, Xi'an, Shaan Xi Province,710032, China
| | - Zhe Wang
- State Key Laboratory of Tumor Biology, Department of Pathology, Xi Jing Hospital, the Fourth Military Medical University, Xi'an, Shaan Xi Province,710032, China.
| | - Shuangping Guo
- State Key Laboratory of Tumor Biology, Department of Pathology, Xi Jing Hospital, the Fourth Military Medical University, Xi'an, Shaan Xi Province,710032, China.
| |
Collapse
|
32
|
Yee LD, Mortimer JE, Natarajan R, Dietze EC, Seewaldt VL. Metabolic Health, Insulin, and Breast Cancer: Why Oncologists Should Care About Insulin. Front Endocrinol (Lausanne) 2020; 11:58. [PMID: 32153503 PMCID: PMC7045050 DOI: 10.3389/fendo.2020.00058] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Accepted: 01/29/2020] [Indexed: 12/13/2022] Open
Abstract
Studies investigating the potential link between adult pre-menopausal obesity [as measured by body mass index (BMI)] and triple-negative breast cancer have been inconsistent. Recent studies show that BMI is not an exact measure of metabolic health; individuals can be obese (BMI > 30 kg/m2) and metabolically healthy or lean (BMI < 25 kg/m2) and metabolically unhealthy. Consequently, there is a need to better understand the molecular signaling pathways that might be activated in individuals that are metabolically unhealthy and how these signaling pathways may drive biologically aggressive breast cancer. One key driver of both type-2 diabetes and cancer is insulin. Insulin is a potent hormone that activates many pathways that drive aggressive breast cancer biology. Here, we review (1) the controversial relationship between obesity and breast cancer, (2) the impact of insulin on organs, subcellular components, and cancer processes, (3) the potential link between insulin-signaling and cancer, and (4) consider time points during breast cancer prevention and treatment where insulin-signaling could be better controlled, with the ultimate goal of improving overall health, optimizing breast cancer prevention, and improving breast cancer survival.
Collapse
|
33
|
Lieb WS, Lungu C, Tamas R, Berreth H, Rathert P, Storz P, Olayioye MA, Hausser A. The GEF-H1/PKD3 signaling pathway promotes the maintenance of triple-negative breast cancer stem cells. Int J Cancer 2019; 146:3423-3434. [PMID: 31745977 DOI: 10.1002/ijc.32798] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 11/05/2019] [Accepted: 11/14/2019] [Indexed: 12/14/2022]
Abstract
Protein kinase D3 (PKD3) is upregulated in triple-negative breast cancer (TNBC) and associated with cell proliferation and metastasis development but its precise pro-oncogenic function is unknown. Here we show that PKD3 is required for the maintenance of the TNBC stem cell population. The depletion of PKD3 in MDA-MB-231 cells reduced the cancer stem cell frequency in vitro and tumor initiation potential in vivo. We further provide evidence that the RhoGEF GEF-H1 is upstream of PKD3 activation in TNBC stem cells. Most importantly, pharmacological PKD inhibition in combination with paclitaxel synergistically decreased oncosphere and colony formation efficiency in vitro and tumor recurrence in vivo. Based on our results we propose that targeting the GEF-H1/PKD3 signaling pathway in combination with chemotherapy might provide an effective therapeutic option for TNBC.
Collapse
Affiliation(s)
- Wolfgang S Lieb
- Institute of Cell Biology and Immunology and Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany
| | - Cristiana Lungu
- Institute of Cell Biology and Immunology and Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany
| | - Raluca Tamas
- Institute of Cell Biology and Immunology and Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany
| | - Hannah Berreth
- Institute of Cell Biology and Immunology and Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany
| | - Philipp Rathert
- Biochemistry Department, Institute of Biochemistry and Technical Biochemistry, University of Stuttgart, Stuttgart, Germany
| | - Peter Storz
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL
| | - Monilola A Olayioye
- Institute of Cell Biology and Immunology and Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany
| | - Angelika Hausser
- Institute of Cell Biology and Immunology and Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany
| |
Collapse
|
34
|
A "NOTCH" Deeper into the Epithelial-To-Mesenchymal Transition (EMT) Program in Breast Cancer. Genes (Basel) 2019; 10:genes10120961. [PMID: 31766724 PMCID: PMC6947643 DOI: 10.3390/genes10120961] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 11/14/2019] [Accepted: 11/18/2019] [Indexed: 12/20/2022] Open
Abstract
Notch signaling is a primitive signaling pathway having various roles in the normal origin and development of each multicellular organisms. Therefore, any aberration in the pathway will inevitably lead to deadly outcomes such as cancer. It has now been more than two decades since Notch was acknowledged as an oncogene in mouse mammary tumor virus-infected mice. Since that discovery, activated Notch signaling and consequent up-regulation of tumor-promoting Notch target genes have been observed in human breast cancer. Moreover, consistent over-expression of Notch ligands and receptors has been shown to correlate with poor prognosis in human breast cancer. Notch regulates a number of key processes during breast carcinogenesis, of which, one key phenomenon is epithelial-mesenchymal transition (EMT). EMT is a key process for large-scale cell movement during morphogenesis at the time of embryonic development. Cancer cells aided by transcription factors usurp this developmental program to execute the multi-step process of tumorigenesis and metastasis. In this review, we recapitulate recent progress in breast cancer research that has provided new perceptions into the molecular mechanisms behind Notch-mediated EMT regulation during breast tumorigenesis.
Collapse
|
35
|
Sandomenico A, Ruvo M. Targeting Nodal and Cripto-1: Perspectives Inside Dual Potential Theranostic Cancer Biomarkers. Curr Med Chem 2019; 26:1994-2050. [PMID: 30207211 DOI: 10.2174/0929867325666180912104707] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 07/13/2018] [Accepted: 07/17/2018] [Indexed: 12/25/2022]
Abstract
BACKGROUND Elucidating the mechanisms of recurrence of embryonic signaling pathways in tumorigenesis has led to the discovery of onco-fetal players which have physiological roles during normal development but result aberrantly re-activated in tumors. In this context, Nodal and Cripto-1 are recognized as onco-developmental factors, which are absent in normal tissues but are overexpressed in several solid tumors where they can serve as theranostic agents. OBJECTIVE To collect, review and discuss the most relevant papers related to the involvement of Nodal and Cripto-1 in the development, progression, recurrence and metastasis of several tumors where they are over-expressed, with a particular attention to their occurrence on the surface of the corresponding sub-populations of cancer stem cells (CSC). RESULTS We have gathered, rationalized and discussed the most interesting findings extracted from some 370 papers related to the involvement of Cripto-1 and Nodal in all tumor types where they have been detected. Data demonstrate the clear connection between Nodal and Cripto-1 presence and their multiple oncogenic activities across different tumors. We have also reviewed and highlighted the potential of targeting Nodal, Cripto-1 and the complexes that they form on the surface of tumor cells, especially of CSC, as an innovative approach to detect and suppress tumors with molecules that block one or more mechanisms that they regulate. CONCLUSION Overall, Nodal and Cripto-1 represent two innovative and effective biomarkers for developing potential theranostic anti-tumor agents that target normal as well as CSC subpopulations and overcome both pharmacological resistance and tumor relapse.
Collapse
Affiliation(s)
- Annamaria Sandomenico
- Istituto di Biostrutture e Bioimmagini, Consiglio Nazionale delle Ricerche (IBB-CNR), via Mezzocannone, 16, 80134, Napoli, Italy
| | - Menotti Ruvo
- Istituto di Biostrutture e Bioimmagini, Consiglio Nazionale delle Ricerche (IBB-CNR), via Mezzocannone, 16, 80134, Napoli, Italy
| |
Collapse
|
36
|
Bonadeo N, Becu-Villalobos D, Cristina C, Lacau-Mengido IM. The Notch system during pubertal development of the bovine mammary gland. Sci Rep 2019; 9:8899. [PMID: 31222104 PMCID: PMC6586787 DOI: 10.1038/s41598-019-45406-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 06/05/2019] [Indexed: 12/27/2022] Open
Abstract
The development of the mammary gland of cows during pre-weaning and puberty will condition its future productive capacity and warrants special study. In this respect, Notch signaling regulates tissue development and fate by modifying cell proliferation and differentiation and has been involved in stem cell maintenance, but has not been extensively studied in the developing mammary glands in cows. We therefore investigated Notch receptor expression and localization, as well as the expression of Notch ligands and target genes in the mammary gland of Holstein heifers in pre- and post-pubertal stages. Notch receptors 1 to 4 were detected by immunohistochemistry in the parenchyma and stroma of the developing gland. The subcellular localization of the four receptors was predominantly cytoplasmic except for NOTCH4, which was mostly nuclear. The membrane and the active intracellular domains of NOTCH paralogues were identified by western blot. NOTCH1 and NOTCH2 active domains increased during pubertal stages while NOTCH3 and NOTCH4 active domains decreased, suggesting strikingly different involvement of NOTCH paralogues in bovine mammary gland development and differentiation. The mRNA expression levels of the target genes HEY1 and HEY2 increased during peri-puberty whereas no variation of HES1 mRNA levels was observed. The mRNA levels of the Notch ligands JAGGED1 and DELTA1 also increased gradually during development. In conclusion, Notch signaling system dynamically varies throughout the development of the mammary gland during puberty pointing to specific time involvement of each component.
Collapse
Affiliation(s)
- Nadia Bonadeo
- Centro de Investigaciones Básicas y Aplicadas, Centro de Investigaciones y Transferencia del Noroeste de la Provincia de Buenos Aires, Monteagudo 2772, Pergamino 2700, Buenos Aires, Argentina
| | - Damasia Becu-Villalobos
- Instituto de Biología y Medicina Experimental, IBYME-CONICET, Vuelta de Obligado 2490, Ciudad Autónoma de Buenos Aires, 1428, Argentina
| | - Carolina Cristina
- Centro de Investigaciones Básicas y Aplicadas, Centro de Investigaciones y Transferencia del Noroeste de la Provincia de Buenos Aires, Monteagudo 2772, Pergamino 2700, Buenos Aires, Argentina
| | - Isabel M Lacau-Mengido
- Instituto de Biología y Medicina Experimental, IBYME-CONICET, Vuelta de Obligado 2490, Ciudad Autónoma de Buenos Aires, 1428, Argentina.
| |
Collapse
|
37
|
Garland MA, Sengupta S, Mathew LK, Truong L, de Jong E, Piersma AH, La Du J, Tanguay RL. Glucocorticoid receptor-dependent induction of cripto-1 ( one-eyed pinhead) inhibits zebrafish caudal fin regeneration. Toxicol Rep 2019; 6:529-537. [PMID: 31249786 PMCID: PMC6584771 DOI: 10.1016/j.toxrep.2019.05.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 05/27/2019] [Accepted: 05/28/2019] [Indexed: 12/15/2022] Open
Abstract
We previously used a chemical genetics approach with the larval zebrafish to identify small molecule inhibitors of tissue regeneration. This led to the discovery that glucocorticoids (GC) block early stages of tissue regeneration by the inappropriate activation of the glucocorticoid receptor (GR). We performed a microarray analysis to identify the changes in gene expression associated with beclomethasone dipropionate (BDP) exposure during epimorphic fin regeneration. Oncofetal cripto-1 showed > eight-fold increased expression in BDP-treated regenerates. We hypothesized that the mis-expression of cripto-1 was essential for BDP to block regeneration. Expression of cripto-1 was not elevated in GR morphants in the presence of BDP indicating that cripto-1 induction was GR-dependent. Partial translational suppression of Cripto-1 in the presence of BDP restored tissue regeneration. Retinoic acid exposure prevented increased cripto-1 expression and permitted regeneration in the presence of BDP. We demonstrated that BDP exposure increased cripto-1 expression in mouse embryonic stem cells and that regulation of cripto-1 by GCs is conserved in mammals.
Collapse
Key Words
- AEC, apical epithelial cap
- BDP, beclomethasone dipropionate
- Beclomethasone dipropionate
- Cripto-1
- DMSO, dimethyl sulfoxide
- EB, embryoid body
- ECM, extracellular matrix
- EMT, epithelial-to-mesenchymal transition
- ERK, extracellular signal-regulated kinase
- Epimorphic regeneration
- FGF, fibroblast growth factor
- GC, glucocorticoid
- GR, glucocorticoid receptor
- Glucocorticoids
- ISH, in situ hybridization
- MIAME, Minimum Information About a Microarray Experiment
- MO, morpholino oligonucleotide
- One-eyed pinhead
- RA, retinoic acid
- SEM, standard error of the mean
- TGF-β, transforming growth factor beta
- Zebrafish
- dpa, days post-amputation
- dpf, days post-fertilization
- eSC, embryonic stem cell
- hpa, hours post-amputation
- hpf, hours post-fertilization
- mLIF, murine leukemia inhibitory factor
- qRT-PCR, quantitative reverse transcription polymerase chain reaction
- zf, zebrafish
Collapse
Affiliation(s)
| | - Sumitra Sengupta
- Department of Environmental and Molecular Toxicology, United States
| | - Lijoy K Mathew
- Department of Environmental and Molecular Toxicology, United States
| | - Lisa Truong
- Department of Environmental and Molecular Toxicology, United States
| | - Esther de Jong
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, the Netherlands.,National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Aldert H Piersma
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, the Netherlands.,National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Jane La Du
- Department of Environmental and Molecular Toxicology, United States
| | - Robert L Tanguay
- Department of Environmental and Molecular Toxicology, United States
| |
Collapse
|
38
|
Hu B, Yan W, Wang M, Cui X, Hu Y, Chen Q, Zhang Y, Qi X, Jiang J. Huaier polysaccharide inhibits the stem-like characteristics of ERα-36 high triple negative breast cancer cells via inactivation of the ERα-36 signaling pathway. Int J Biol Sci 2019; 15:1358-1367. [PMID: 31337967 PMCID: PMC6643138 DOI: 10.7150/ijbs.27360] [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: 05/19/2018] [Accepted: 04/14/2019] [Indexed: 01/05/2023] Open
Abstract
Triple negative breast cancer (TNBC) is a highly aggressive cancer and lack of targeting therapies. It is believed that the breast cancer stem cells (BCSCs) are responsible for the aggressive characteristics of TNBC. Hence, developing BCSC-targeting agents may provide new therapeutic strategies for the patients. Huaier polysaccharide (HP), an active ingredient extracted from the mushroom Trametes robiniophila Murr, has been widely used in clinical anti-cancer treatments in China. Here we demonstrated that HP could target BCSCs in TNBC cells, resulting in decreased mammosphere formation, downregulated expression of stem-related genes and reduced proportion of aldehyde dehydrogenase positive cells in vitro, and inhibited xenograft tumor formation in vivo. Mechanically, HP markedly reduced the expression of estrogen receptor α-36 (ERα-36), a recently identified subtype of estrogen receptor α, and attenuated ERα-36-mediated activation of AKT/β-catenin signaling in ERα-36high TNBC cells. This study provides a new insight into the mechanism of HP on BCSC-targeting therapy and new ideas for comprehensive treatment strategies for TNBC.
Collapse
Affiliation(s)
- Baoquan Hu
- Breast Disease Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Wenting Yan
- Breast Disease Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Minghao Wang
- Breast Disease Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Xiang Cui
- Breast Disease Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Ying Hu
- Breast Disease Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Qingqiu Chen
- Breast Disease Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Yi Zhang
- Breast Disease Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Xiaowei Qi
- Breast Disease Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Jun Jiang
- Breast Disease Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| |
Collapse
|
39
|
In vivo and in vitro study of co-expression of LMP1 and Cripto-1 in nasopharyngeal carcinoma. Braz J Otorhinolaryngol 2019; 86:617-625. [PMID: 31375471 PMCID: PMC9422379 DOI: 10.1016/j.bjorl.2019.04.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 04/07/2019] [Indexed: 12/20/2022] Open
Abstract
INTRODUCTION Nasopharyngeal carcinoma, an epithelial-derived malignant tumor which because of its anatomical location and atypical early symptoms, when diagnosed invasion and metastasis often have occurred. This requires a better understanding of the development mechanism, identifying diagnostic markers, and developing new treatment strategies. OBJECTIVE To study the relationship of LMP1 and Cripto-1 in nasopharyngeal carcinoma. METHODS The expression of LMP1 and Cripto-1 in specimens obtained from nasopharyngeal carcinoma patients (n=42) and nasopharyngitis patients (n=22) were examined. The expression of LMP1 and Cripto-1 in LMP1-negative and LMP1-positive (CNE1-LMP1) cells were also examined. RESULTS The expression of LMP1 and Cripto-1 was significantly higher in nasopharyngeal carcinoma than in nasopharyngitis (p<0.05). Their expression in nasopharyngeal carcinoma with metastasis were significantly higher than that without metastasis (p<0.05), which was correlated with TNM staging (p<0.05). High Cripto-1 expression and high proliferation rate were seen in CNE1-LMP1 cells. CONCLUSIONS The expression of LMP1 and Cripto-1 in nasopharyngeal carcinoma is positively related. Their co-expression might contribute to the proliferation and metastasis of nasopharyngeal carcinoma.
Collapse
|
40
|
Wang Z, Li Y, Xiao Y, Lin HP, Yang P, Humphries B, Gao T, Yang C. Integrin α9 depletion promotes β-catenin degradation to suppress triple-negative breast cancer tumor growth and metastasis. Int J Cancer 2019; 145:2767-2780. [PMID: 31008533 DOI: 10.1002/ijc.32359] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 03/26/2019] [Accepted: 04/16/2019] [Indexed: 12/27/2022]
Abstract
Although integrin α9 (ITGA9) is known to be involved in cell adhesion and motility, its expression in cancer and its role in tumor growth and metastasis remain largely unknown. Our study was designed to investigate the role of ITGA9 in triple-negative breast cancer (TNBC). ITGA9 expression in TNBC cells was knocked out (KO) using CRISPR/Cas9 technology. Four orthotopic mouse mammary xenograft tumor models coupled with cell culture studies were performed to determine the effect of ITGA9 depletion on TNBC tumor growth and metastasis and the underlying mechanism. Bioinformatics analysis showed that ITGA9 level is significantly higher in TNBC than other breast cancer subtypes, and higher ITGA9 level is associated with significantly worse distant metastasis-free survival and recurrence-free survival in TNBC patients. Experimentally, ITGA9 KO significantly reduced TNBC cell cancer stem cell (CSC)-like property, tumor angiogenesis, tumor growth and metastasis by promoting β-catenin degradation. Further mechanistic studies revealed that ITGA9 KO causes integrin-linked kinase (ILK) relocation from the membrane region to the cytoplasm, where it interacts with protein kinase A (PKA) and inhibits PKA activity leading to increased activity of glycogen synthase kinase 3 (GSK3) and subsequent β-catenin degradation. Overexpressing β-catenin in ITGA9 KO cells reversed the inhibitory effect of ITGA9 KO on tumor growth and metastasis. Furthermore, ITGA9 downregulation in TNBC tumors by nanoparticle-mediated delivery of ITGA9 siRNA drastically decreased tumor angiogenesis, tumor growth and metastasis. These findings indicate that ITGA9 depletion suppresses TNBC tumor growth and metastasis by promoting β-catenin degradation through the ILK/PKA/GSK3 pathway.
Collapse
Affiliation(s)
- Zhishan Wang
- Department of Toxicology and Cancer Biology, and Center for Research on Environment Disease, College of Medicine, University of Kentucky, Lexington, KY
| | - Yunfei Li
- Department of Toxicology and Cancer Biology, and Center for Research on Environment Disease, College of Medicine, University of Kentucky, Lexington, KY
| | - Yajuan Xiao
- Department of Toxicology and Cancer Biology, and Center for Research on Environment Disease, College of Medicine, University of Kentucky, Lexington, KY.,Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Hsuan-Pei Lin
- Department of Toxicology and Cancer Biology, and Center for Research on Environment Disease, College of Medicine, University of Kentucky, Lexington, KY
| | - Ping Yang
- Department of Toxicology and Cancer Biology, and Center for Research on Environment Disease, College of Medicine, University of Kentucky, Lexington, KY.,School of Public Health, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Brock Humphries
- Department of Radiology, University of Michigan, Ann Arbor, MI
| | - Tianyan Gao
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, KY
| | - Chengfeng Yang
- Department of Toxicology and Cancer Biology, and Center for Research on Environment Disease, College of Medicine, University of Kentucky, Lexington, KY
| |
Collapse
|
41
|
López-Ozuna VM, Hachim IY, Hachim MY, Lebrun JJ, Ali S. Prolactin modulates TNBC aggressive phenotype limiting tumorigenesis. Endocr Relat Cancer 2019; 26:321-337. [PMID: 30640712 DOI: 10.1530/erc-18-0523] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 01/14/2019] [Indexed: 12/21/2022]
Abstract
Triple-negative breast cancer (TNBC) accounts for ~20% of all breast cancer cases. The management of TNBC represents a challenge due to its aggressive phenotype, heterogeneity and lack of targeted therapy. Loss of cell differentiation and enrichment with breast cancer stem-like cells (BCSC) are features of TNBC contributing to its aggressive nature. Here, we found that treatment of TNBC cells with PRL significantly depletes the highly tumorigenic BCSC subpopulations CD44+/CD24- and ALDH+ and differentiates them to the least tumorigenic CD44-/CD24- and ALDH- phenotype with limited tumorsphere formation and self-renewal capacities. Importantly, we found PRL to induce a heterochromatin phenotype marked by histone H3 lysine 9 trimethylation (H3K9me3) and accompanied by ultra-structural cellular architecture associated with differentiation and senescence rendering the cells refractory to growth signals. Crucially, we found PRL to mediate these effects in vivo in a pre-clinical animal xenograft of TNBC controlling tumor growth. These results reveal that the lactogenic hormone PRL may exert its anti-tumorigenic effects on TNBC through cellular reprogramming indicative of differentiation resulting in the depletion of BCSCs and restricting tumorigenesis.
Collapse
Affiliation(s)
- Vanessa M López-Ozuna
- Department of Medicine, Cancer Research Program, McGill University Health Centre, McGill University, Montreal, Québec, Canada
| | - Ibrahim Y Hachim
- Department of Medicine, Cancer Research Program, McGill University Health Centre, McGill University, Montreal, Québec, Canada
| | - Mahmood Y Hachim
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Jean-Jacques Lebrun
- Department of Medicine, Cancer Research Program, McGill University Health Centre, McGill University, Montreal, Québec, Canada
| | - Suhad Ali
- Department of Medicine, Cancer Research Program, McGill University Health Centre, McGill University, Montreal, Québec, Canada
| |
Collapse
|
42
|
Focà G, Iaccarino E, Focà A, Sanguigno L, Untiveros G, Cuevas-Nunez M, Strizzi L, Leonardi A, Ruvo M, Sandomenico A. Development of conformational antibodies targeting Cripto-1 with neutralizing effects in vitro. Biochimie 2019; 158:246-256. [PMID: 30703478 DOI: 10.1016/j.biochi.2019.01.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 01/22/2019] [Indexed: 01/14/2023]
Abstract
Human Cripto-1 (Cripto-1), the founding member of the EGF-CFC superfamily, is a key regulator of many processes during embryonic development and oncogenesis. Cripto-1 is barely present or even absent in normal adult tissues while it is aberrantly re-expressed in various tumors. Blockade of the CFC domain-mediated Cripto-1 functions is acknowledged as a promising therapeutic intervention point to inhibit the tumorigenic activity of the protein. In this work, we report the generation and characterization of murine monoclonal antibodies raised against the synthetic folded CFC [112-150] domain of the human protein. Through subtractive ELISA assays clones were screened for the ability to specifically recognize "hot spot" residues on the CFC domain, which are crucial for the interaction with Activin Type I receptor (ALK4) and GRP78. On selected antibodies, SPR and epitope mapping studies have confirmed their specificity and have revealed that recognition occurs only on a conformational epitope. Furthermore, FACS analyses have confirmed the ability of 1B4 antibody to recognize the membrane-anchored and soluble native Cripto-1 protein in a panel of human cancer cells. Finally, we have evaluated its functional effects through in vitro cellular signaling assays and cell cycle analysis. These findings suggest that the selected anti-CFC mAbs have the potential to neutralize the protein oncogenic activity and may be used as theranostic molecules suitable as tumor homing agents for Cripto-1-overexpressing cancer cells and tissues and to overcome drug-resistance in routine cancer therapies.
Collapse
Affiliation(s)
- Giuseppina Focà
- Institute of Biostructure and Bioimaging, National Research Council (IBB-CNR), Naples, Italy
| | - Emanuela Iaccarino
- Institute of Biostructure and Bioimaging, National Research Council (IBB-CNR), Naples, Italy
| | - Annalia Focà
- Institute of Biostructure and Bioimaging, National Research Council (IBB-CNR), Naples, Italy
| | - Luca Sanguigno
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy
| | - Gustavo Untiveros
- Midwestern University, Colleges of Graduate Studies, Dwners Grove, Chicago, IL, USA
| | - Maria Cuevas-Nunez
- Midwestern University, Colleges of Graduate Studies, Dwners Grove, Chicago, IL, USA; College of Dental Medicine, Dwners Grove, Chicago, IL, USA
| | - Luigi Strizzi
- Midwestern University, Colleges of Graduate Studies, Dwners Grove, Chicago, IL, USA
| | - Antonio Leonardi
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy
| | - Menotti Ruvo
- Institute of Biostructure and Bioimaging, National Research Council (IBB-CNR), Naples, Italy.
| | - Annamaria Sandomenico
- Institute of Biostructure and Bioimaging, National Research Council (IBB-CNR), Naples, Italy.
| |
Collapse
|
43
|
Castro NP, Rangel MC, Merchant AS, MacKinnon G, Cuttitta F, Salomon DS, Kim YS. Sulforaphane Suppresses the Growth of Triple-negative Breast Cancer Stem-like Cells In vitro and In vivo. Cancer Prev Res (Phila) 2019; 12:147-158. [PMID: 30679159 DOI: 10.1158/1940-6207.capr-18-0241] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 11/20/2018] [Accepted: 01/11/2019] [Indexed: 12/21/2022]
Abstract
Triple-negative breast cancer (TNBC) represents the poorest prognosis among all of breast cancer subtypes with no currently available effective therapy. In this study, we hypothesized that sulforaphane, a dietary component abundant in broccoli and its sprouts, can inhibit malignant cell proliferation and tumor sphere formation of cancer stem-like cells (CSC) in TNBC. CSC population was isolated using FACS analysis with the combined stem cell surface markers, CD44+/CD24-/CD49f+ The effect of sulforaphane on a stem-related embryonic oncogene CRIPTO-1/TDGF1 (CR1) was evaluated via ELISA. In vivo, BalbC/nude mice were supplemented with sulforaphane before and after TNBC cell inoculation (daily intraperitoneal injection of 50 mg sulforaphane/kg for 5 and 3 weeks, respectively), and the effects of sulforaphane during mammary tumor initiation and growth were accessed with NanoString gene analysis. We found that sulforaphane can inhibit cell proliferation and mammosphere formation of CSCs in TNBC. Further analysis of gene expression in these TNBC tumor cells revealed that sulforaphane significantly decreases the expression of cancer-specific CR1, CRIPTO-3/TDGF1P3 (CR3, a homologue of CR1), and various stem cell markers including Nanog, aldehyde dehydrogenase 1A1 (ALDH1A1), Wnt3, and Notch4. Our results suggest that sulforaphane may control the malignant proliferation of CSCs in TNBC via Cripto-mediated pathway by either suppressing its expression and/or by inhibiting Cripto/Alk4 protein complex formation. Thus, the use of sulforaphane for chemoprevention of TNBC is plausible and warrants further clinical evaluation.
Collapse
Affiliation(s)
- Nadia P Castro
- Tumor Growth Factor Section, Mouse Cancer Genetics Program, NCI, Frederick, Maryland
| | - Maria C Rangel
- Tumor Growth Factor Section, Mouse Cancer Genetics Program, NCI, Frederick, Maryland
| | - Anand S Merchant
- Center for Cancer Research Collaborative Bioinformatics Core, NCI, Bethesda, Maryland
| | - Gabriel MacKinnon
- Tumor Growth Factor Section, Mouse Cancer Genetics Program, NCI, Frederick, Maryland
| | - Frank Cuttitta
- Tumor Growth Factor Section, Mouse Cancer Genetics Program, NCI, Frederick, Maryland
| | - David S Salomon
- Tumor Growth Factor Section, Mouse Cancer Genetics Program, NCI, Frederick, Maryland
| | - Young S Kim
- Nutritional Science Research Group, Division of Cancer Prevention, NCI, Rockville, Maryland.
| |
Collapse
|
44
|
Elevated leptin disrupts epithelial polarity and promotes premalignant alterations in the mammary gland. Oncogene 2019; 38:3855-3870. [PMID: 30670780 PMCID: PMC6525037 DOI: 10.1038/s41388-019-0687-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 12/04/2018] [Accepted: 12/18/2018] [Indexed: 12/15/2022]
Abstract
Obesity is a highly prevalent and modifiable breast cancer risk factor. While the role of obesity in fueling breast cancer progression is well established, the mechanisms linking obesity to breast cancer initiation are poorly understood. A hallmark of breast cancer initiation is the disruption of apical polarity in mammary glands. Here we show that mice with diet-induced obesity display mislocalization of Par3, a regulator of cellular junctional complexes defining mammary epithelial polarity. We found that epithelial polarity loss also occurs in a 3D coculture system that combines acini with human mammary adipose tissue, and establish that a paracrine effect of the tissue adipokine leptin causes loss of polarity by overactivation of the PI3K/Akt pathway. Leptin sensitizes non-neoplastic cells to proliferative stimuli, causes mitotic spindle misalignment, and expands the pool of cells with stem/progenitor characteristics, which are early steps for cancer initiation. We also found that normal breast tissue samples with high leptin/adiponectin transcript ratio characteristic of obesity have an altered distribution of apical polarity markers. This effect is associated with increased epithelial cell layers. Our results provide a molecular basis for early alterations in epithelial architecture during obesity-mediated cancer initiation.
Collapse
|
45
|
Li Y, Xiao Y, Lin HP, Reichel D, Bae Y, Lee EY, Jiang Y, Huang X, Yang C, Wang Z. In vivo β-catenin attenuation by the integrin α5-targeting nano-delivery strategy suppresses triple negative breast cancer stemness and metastasis. Biomaterials 2019; 188:160-172. [DOI: 10.1016/j.biomaterials.2018.10.019] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 10/16/2018] [Accepted: 10/17/2018] [Indexed: 12/13/2022]
|
46
|
Bao B, Prasad AS. Targeting CSC in a Most Aggressive Subtype of Breast Cancer TNBC. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1152:311-334. [DOI: 10.1007/978-3-030-20301-6_17] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
47
|
Wen XF, Chen M, Wu Y, Chen MN, Glogowska A, Klonisch T, Zhang GJ. Inhibitor of DNA Binding 2 Inhibits Epithelial-Mesenchymal Transition via Up-Regulation of Notch3 in Breast Cancer. Transl Oncol 2018; 11:1259-1270. [PMID: 30119050 PMCID: PMC6097462 DOI: 10.1016/j.tranon.2018.07.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 07/23/2018] [Indexed: 02/05/2023] Open
Abstract
Breast cancer is the second leading cause of cancer death in women worldwide. Incurable metastatic breast disease presents a major clinical challenge and is the main cause of breast cancer-related death. The epithelial-mesenchymal transition (EMT) is a critical early promoter of metastasis. In the present study, we identified a novel role for the inhibitor of DNA binding 2 (Id2), a member of the basic helix-loop-helix protein family, during the EMT of breast cancer. Expression of Id2 was positively correlated with Notch3 in breast cancer cells. Low expression of Id2 and Notch3 was associated with worse distant metastasis-free survival in breast cancer patients. The present study revealed that Id2 activated Notch3 expression by blocking E2A binding to an E-box motif in the Notch3 promoter. The Id2-mediated up-regulation of Notch3 expression at both the mRNA and protein levels resulted in an attenuated EMT, which was associated with reduced motility and matrix invasion of ER-positive and -negative human breast cancer cells and the emergence of E-cadherin expression and reduction in the mesenchymal marker vimentin in triple-negative breast cancer cells. In summary, our findings identified Id2 as a suppressor of the EMT and positive transcriptional regulator of Notch3 in breast cancer. Id2 and Notch3 may serve as novel prognostic markers in a subpopulation of ER-positive breast cancer patients.
Collapse
Affiliation(s)
- Xiao-Fen Wen
- Department of Breast Medical Oncology, Cancer Hospital of Shantou University Medical College, 7 Raoping Road, Shantou, China; ChangJiang Scholar's Laboratory, Shantou University Medical College, 22 Xinling Road, Shantou, China
| | - Min Chen
- ChangJiang Scholar's Laboratory, Shantou University Medical College, 22 Xinling Road, Shantou, China; Xiang'an Hospital, Xiamen University, 2000 East Xiang'an Rd, Xiamen, Fujian, China
| | - Yang Wu
- ChangJiang Scholar's Laboratory, Shantou University Medical College, 22 Xinling Road, Shantou, China
| | - Min-Na Chen
- Department of Breast Medical Oncology, Cancer Hospital of Shantou University Medical College, 7 Raoping Road, Shantou, China; ChangJiang Scholar's Laboratory, Shantou University Medical College, 22 Xinling Road, Shantou, China
| | - Aleksandra Glogowska
- Dept. of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Thomas Klonisch
- Dept. of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada.
| | - Guo-Jun Zhang
- ChangJiang Scholar's Laboratory, Shantou University Medical College, 22 Xinling Road, Shantou, China; Xiang'an Hospital, Xiamen University, 2000 East Xiang'an Rd, Xiamen, Fujian, China.
| |
Collapse
|
48
|
Bemanian V, Noone JC, Sauer T, Touma J, Vetvik K, Søderberg-Naucler C, Lindstrøm JC, Bukholm IR, Kristensen VN, Geisler J. Somatic EP300-G211S mutations are associated with overall somatic mutational patterns and breast cancer specific survival in triple-negative breast cancer. Breast Cancer Res Treat 2018; 172:339-351. [PMID: 30132219 DOI: 10.1007/s10549-018-4927-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 08/17/2018] [Indexed: 10/28/2022]
Abstract
PURPOSE We have compared the mutational profiles of human breast cancer tumor samples belonging to all major subgroups with special emphasis on triple-negative breast cancer (TNBC). Our major goal was to identify specific mutations that could be potentially used for clinical decision making in TNBC patients. PATIENTS AND METHODS Primary tumor specimens from 149 Norwegian breast cancer patients were available. We analyzed the tissue samples for somatic mutations in 44 relevant breast cancer genes by targeted next-generation sequencing. As a second confirmatory technique, we performed pyrosequencing on selected samples. RESULTS We observed a distinct subgroup of TNBC patients, characterized by an almost completely lack of pathogenic somatic mutations. A point mutation in the adenoviral E1A binding protein p300 (EP300-G211S) was significantly correlated to this TNBC subgroup. The EP300-G211S mutation was exclusively found in the TNBC patients and its presence reduced the chance for other pathological somatic mutations in typical breast cancer genes investigated in our gene panel by 94.9% (P < 0.005). Interestingly, the EP300-G211S mutation also predicted a lower risk for relapses and decreased breast cancer-specific mortality during long-term follow-up of the patients. CONCLUSION Next-generation sequencing revealed specific mutations in EP300 to be associated with the mutational patterns in typical breast cancer genes and long-term outcome of triple-negative breast cancer patients.
Collapse
Affiliation(s)
- Vahid Bemanian
- Section of Gene Technology, Akershus University Hospital, 1478, Lørenskog, Norway
| | | | - Torill Sauer
- Department of Pathology, Akershus University Hospital, 1478, Lørenskog, Norway.,Institute of Clinical Medicine, University of Oslo, Campus at Akershus University Hospital, 1478, Lørenskog, Norway
| | - Joel Touma
- Department of Breast and Endocrine Surgery, Akershus University Hospital, 1478, Lørenskog, Norway.,Department of Oncology, Akershus University Hospital, 1478, Lørenskog, Norway
| | - Katja Vetvik
- Institute of Clinical Medicine, University of Oslo, Campus at Akershus University Hospital, 1478, Lørenskog, Norway.,Department of Breast and Endocrine Surgery, Akershus University Hospital, 1478, Lørenskog, Norway
| | - Cecilia Søderberg-Naucler
- Department of Medicine at Solna, Experimental Cardiovascular Research Unit and Departments of Medicine and Neurology, Center for Molecular Medicine, Karolinska Institute, 17176, Stockholm, Sweden
| | - Jonas Christoffer Lindstrøm
- Institute of Clinical Medicine, University of Oslo, Campus at Akershus University Hospital, 1478, Lørenskog, Norway.,Health Services Research Unit, Akershus University Hospital, 1478, Lørenskog, Norway
| | - Ida Rashida Bukholm
- Department of Breast and Endocrine Surgery, Akershus University Hospital, 1478, Lørenskog, Norway.,Norwegian System of Compensation to Patients, Oslo, Norway.,The Norwegian University of Life Sciences, Ås, Norway
| | - Vessela N Kristensen
- Institute of Clinical Medicine, University of Oslo, Campus at Akershus University Hospital, 1478, Lørenskog, Norway.,Clinical Molecular Biology (EPIGEN), Akershus University Hospital, 1478, Lørenskog, Norway
| | - Jürgen Geisler
- Institute of Clinical Medicine, University of Oslo, Campus at Akershus University Hospital, 1478, Lørenskog, Norway. .,Department of Oncology, Akershus University Hospital, 1478, Lørenskog, Norway.
| |
Collapse
|
49
|
Zhao Q, Zhang X, Cai H, Zhang P, Kong D, Ge X, Du M, Liang R, Dong W. Anticancer effects of plant derived Anacardic acid on human breast cancer MDA-MB-231 cells. Am J Transl Res 2018; 10:2424-2434. [PMID: 30210681 PMCID: PMC6129544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 07/31/2018] [Indexed: 06/08/2023]
Abstract
Triple negative breast cancer (TNBC) accounts for about 10-15% of all breast cancers. It is a heterogeneous disease, characterized by early relapse, aggressive behavior, and poor prognosis, when compared to other breast cancer subtypes. Interestingly, most of the heat shock protein 90 (Hsp90) client proteins are oncoproteins, and some are closely related to the key factors that promote the progression of TNBC. Anacardic acid (AA), which is commonly seen in natural plants of Anacardiaceae, exhibits potent Hsp90 ATPase inhibition activity. In this study, the anticancer effects of AA on TNBC MDA-MB-231 cells were investigated. The results of our study showed that AA inhibited cell proliferation, induced G0/G1-phase cell cycle arrest, suppressed cell invasion and migration, and induced apoptosis in the MDA-MB-231 cells. Regulation of the key Hsp90-dependent tumor-related molecules or endoplasmic reticulum stress (ERS) related molecules, such as GRP78, Hsp70, CDK-4, MMP-9, Bcl-2, and Mcl-1 by AA may be related to these effects. Taken together, our results suggest that AA shows potential as a possible new drug for therapy of TNBC.
Collapse
Affiliation(s)
- Qing Zhao
- Department of Pharmacy, Affiliated Hospital of Jiangnan UniversityWuxi, Jiangsu, People’s Republic of China
| | - Xiaofeng Zhang
- Department of Respiration, Affiliated Hospital of Jiangnan UniversityWuxi, Jiangsu, People’s Republic of China
| | - Haifeng Cai
- Department of Pharmacy, The Fifth People’s Hospital of WuxiWuxi, Jiangsu, People’s Republic of China
| | - Pei Zhang
- Department of Pharmacy, Bengbu Medical CollegeAnhui, People’s Republic of China
| | - Dong Kong
- Department of Radiotherapy, Affiliated Hospital of Jiangnan UniversityWuxi, Jiangsu, People’s Republic of China
| | - Xiaosong Ge
- Department of Oncology, Affiliated Hospital of Jiangnan UniversityWuxi, Jiangsu, People’s Republic of China
| | - Min Du
- Department of Pharmacy, Affiliated Hospital of Jiangnan UniversityWuxi, Jiangsu, People’s Republic of China
| | - Rong Liang
- School of Chemical and Material Engineering, Jiangnan UniversityWuxi, Jiangsu, People’s Republic of China
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, Jiangnan UniversityWuxi, Jiangsu, People’s Republic of China
| | - Wenxia Dong
- Department of Education and Nephrology, Affiliated Hospital of Jiangnan UniversityWuxi, Jiangsu, People’s Republic of China
| |
Collapse
|
50
|
Mei L, He L, Song Y, Lv Y, Zhang L, Hao F, Xu M. Association between obesity with disease-free survival and overall survival in triple-negative breast cancer: A meta-analysis. Medicine (Baltimore) 2018; 97:e0719. [PMID: 29742734 PMCID: PMC5959383 DOI: 10.1097/md.0000000000010719] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE To investigate the relationship between obesity and disease-free survival (DFS) and overall survival (OS) of triple-negative breast cancer. METHODS Citations were searched in PubMed, Cochrane Library, and Web of Science. Random effect model meta-analysis was conducted by using Revman software version 5.0, and publication bias was evaluated by creating Egger regression with STATA software version 12. RESULTS Nine studies (4412 patients) were included for DFS meta-analysis, 8 studies (4392 patients) include for OS meta-analysis. There were no statistical significances between obesity with DFS (P = .60) and OS (P = .71) in triple-negative breast cancer (TNBC) patients. CONCLUSION Obesity has no impact on DFS and OS in patients with TNBC.
Collapse
Affiliation(s)
- Lin Mei
- Department of Oncology, The PLA Navy Anqing Hospital, Anqing, Anhui Province. Breast Center B Ward, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province Department of Gastroenterology, The Second Hospital of Anhui Medical University, Hefei, Anhui Province Department of Anesthesiology Department of Rehabilitation Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, China
| | | | | | | | | | | | | |
Collapse
|