101
|
Zhu Y, Yu F, Jiao Y, Feng J, Tang W, Yao H, Gong C, Chen J, Su F, Zhang Y, Song E. Reduced miR-128 in breast tumor-initiating cells induces chemotherapeutic resistance via Bmi-1 and ABCC5. Clin Cancer Res 2011; 17:7105-15. [PMID: 21953503 DOI: 10.1158/1078-0432.ccr-11-0071] [Citation(s) in RCA: 197] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE Tumor-initiating cells are resistant to chemotherapy, but how microRNAs play a role in regulating drug resistance of breast tumor-initiating cells (BT-IC) needs to be clarified. EXPERIMENTAL DESIGN Lentivirus-mediated miR-128 transduction was done in BT-ICs, enriched by mammosphere cultures or CD44(+)CD24(-) fluorescence-activated cell sorting. Apoptosis and DNA damage were determined upon treatment with doxorubicin. Expression of miR-128 in breast cancer tissues was examined by in situ hybridization and correlated with breast tumor response to neoadjuvant chemotherapy and patient survival. RESULTS MiR-128 was significantly reduced in chemoresistant BT-ICs enriched from breast cancer cell lines and primary breast tumors (P < 0.01), accompanied by an overexpression of Bmi-1 and ABCC5, which were identified as targets of miR-128. Ectopic expression of miR-128 reduced the protein levels of Bmi-1 and ABCC5 in BT-ICs, along with decreased cell viability (P < 0.001) and increased apoptosis (P < 0.001) and DNA damage (P < 0.001) in the presence of doxorubicin. Reduced miR-128 expression in breast tumor tissues was associated with chemotherapeutic resistance (P < 0.001) and poor survival of breast cancer patients (P < 0.05; n = 57). CONCLUSIONS Reduction in miR-128 leading to Bmi-1 and ABCC5 overexpression is a stem cell-like feature of BT-ICs, which contributes to chemotherapeutic resistance in breast cancers. Ectopic expression of miR-128 sensitizes BT-ICs to the proapoptotic and DNA-damaging effects of doxorubicin, indicating therapeutic potential.
Collapse
Affiliation(s)
- Yinghua Zhu
- School of Life Sciences, Sun-Yat-Sen University, Guangzhou, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
102
|
Ai J, Tang Q, Wu Y, Xu Y, Feng T, Zhou R, Chen Y, Gao X, Zhu Q, Yue X, Pan Q, Xu S, Li J, Huang M, Daugherty-Holtrop J, He Y, Xu HE, Fan J, Ding J, Geng M. The role of polymeric immunoglobulin receptor in inflammation-induced tumor metastasis of human hepatocellular carcinoma. J Natl Cancer Inst 2011; 103:1696-712. [PMID: 22025622 PMCID: PMC3216966 DOI: 10.1093/jnci/djr360] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Background Expression of the polymeric immunoglobulin receptor (pIgR), a transporter of polymeric IgA and IgM, is commonly increased in response to viral or bacterial infections, linking innate and adaptive immunity. Abnormal expression of pIgR in cancer was also observed, but its clinical relevance remains uncertain. Methods A human hepatocellular carcinoma (HCC) tissue microarray (n = 254) was used to investigate the association between pIgR expression and early recurrence. An experimental lung metastasis model using severe combined immune-deficient mice was applied to determine the metastatic potential of Madin–Darby canine kidney (n = 5 mice per group) and SMMC-7721 (n = 12 mice per group) cells overexpressing pIgR vs control cells. RNA interference, immunoprecipitation, and immunoblotting were performed to investigate the potential role for pIgR in the induction of epithelial–mesenchymal transition (EMT). In vitro studies (co-immunoprecipitation, immunoblotting, and migration, invasion, and adhesion assays) were used to determine the mechanisms behind pIgR-mediated metastasis. All statistical tests were two-sided. Results High expression of pIgR was statistically significantly associated with early recurrence in early-stage HCC and in hepatitis B surface antigen–positive HCC patients (log-rank P = .02). Mice injected with pIgR-overexpressing cells had a statistically significantly higher number of lung metastases compared with respective control cells (Madin–Darby canine kidney cells: pIgR mean = 29.4 metastatic nodules per lung vs control mean = 0.0 metastatic nodules per lung, difference = 29.4 metastatic nodules per lung, 95% confidence interval = 13.0 to 45.8, P = .001; SMMC-7721 cells: pIgR mean = 10.4 metastatic nodules per lung vs control mean = 2.2 metastatic nodules per lung, difference = 8.2 metastatic nodules per lung, 95% confidence interval = 1.0 to 15.5, P = .03). Furthermore, high expression of pIgR was sufficient to induce EMT through activation of Smad signaling. Conclusions pIgR plays a role in the induction of EMT. Our results identify pIgR as a potential link between hepatitis B virus–derived hepatitis and HCC metastasis and provide evidence in support of pIgR as a prognostic biomarker for HCC and a potential therapeutic target.
Collapse
Affiliation(s)
- Jing Ai
- Division of Anti-tumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Rd., Shanghai, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
103
|
Wilson BJ, Schatton T, Zhan Q, Gasser M, Ma J, Saab KR, Schanche R, Waaga-Gasser AM, Gold JS, Huang Q, Murphy GF, Frank MH, Frank NY. ABCB5 identifies a therapy-refractory tumor cell population in colorectal cancer patients. Cancer Res 2011; 71:5307-16. [PMID: 21652540 PMCID: PMC3395026 DOI: 10.1158/0008-5472.can-11-0221] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Identification and reversal of treatment resistance mechanisms of clinically refractory tumor cells is critical for successful cancer therapy. Here we show that ATP-binding cassette member B5 (ABCB5) identifies therapy-refractory tumor cells in colorectal cancer patients following fluorouracil (5-FU)-based chemoradiation therapy and provide evidence for a functional role of ABCB5 in colorectal cancer 5-FU resistance. Examination of human colon and colorectal cancer specimens revealed ABCB5 to be expressed only on rare cells within healthy intestinal tissue, whereas clinical colorectal cancers exhibited substantially increased levels of ABCB5 expression. Analysis of successive, patient-matched biopsy specimens obtained prior to and following neoadjuvant 5-FU-based chemoradiation therapy in a series of colorectal cancer patients revealed markedly enhanced abundance of ABCB5-positive tumor cells when residual disease was detected. Consistent with this finding, the ABCB5-expressing tumor cell population was also treatment refractory and exhibited resistance to 5-FU-induced apoptosis in a colorectal cancer xenograft model of 5-FU monotherapy. Mechanistically, short hairpin RNA-mediated ABCB5 knockdown significantly inhibited tumorigenic xenograft growth and sensitized colorectal cancer cells to 5-FU-induced cell killing. Our results identify ABCB5 as a novel molecular marker of therapy-refractory tumor cells in colorectal cancer patients and point to a need for consistent eradication of ABCB5-positive resistant tumor cell populations for more effective colorectal cancer therapy.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B
- ATP Binding Cassette Transporter, Subfamily B, Member 1/analysis
- ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors
- ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology
- Adenocarcinoma/chemistry
- Adenocarcinoma/drug therapy
- Adenocarcinoma/pathology
- Adenocarcinoma/radiotherapy
- Adenocarcinoma/surgery
- Animals
- Antimetabolites, Antineoplastic/administration & dosage
- Antimetabolites, Antineoplastic/pharmacology
- Antimetabolites, Antineoplastic/therapeutic use
- Biomarkers, Tumor/analysis
- Colorectal Neoplasms/chemistry
- Colorectal Neoplasms/drug therapy
- Colorectal Neoplasms/pathology
- Colorectal Neoplasms/radiotherapy
- Colorectal Neoplasms/surgery
- Combined Modality Therapy
- Drug Resistance, Neoplasm/physiology
- Fluorouracil/administration & dosage
- Fluorouracil/pharmacology
- Fluorouracil/therapeutic use
- Humans
- Interleukin Receptor Common gamma Subunit/deficiency
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Neoadjuvant Therapy
- Neoplasm Proteins/analysis
- Neoplasm Proteins/antagonists & inhibitors
- Neoplasm Proteins/physiology
- RNA, Small Interfering/pharmacology
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Brian J. Wilson
- Transplantation Research Center, Children’s Hospital Boston, Boston, MA
| | - Tobias Schatton
- Transplantation Research Center, Children’s Hospital Boston, Boston, MA
| | - Qian Zhan
- Department of Pathology, Brigham & Women’s Hospital, Boston, MA
| | - Martin Gasser
- Department of Surgery, University of Würzburg, Würzburg, Germany
| | - Jie Ma
- Transplantation Research Center, Children’s Hospital Boston, Boston, MA
| | - Karim R. Saab
- Transplantation Research Center, Children’s Hospital Boston, Boston, MA
| | - Robin Schanche
- Department of Pathology, Brigham & Women’s Hospital, Boston, MA
| | | | - Jason S. Gold
- Surgical Service, VA Boston Healthcare System, Boston, MA
- Department of Surgery, Brigham & Women’s Hospital, Boston, MA
| | - Qin Huang
- Department of Pathology, VA Boston Healthcare System, Boston, MA
| | | | - Markus H. Frank
- Transplantation Research Center, Children’s Hospital Boston, Boston, MA
| | - Natasha Y. Frank
- Department of Medicine, VA Boston Healthcare System, Boston, MA
- Division of Genetics, Brigham & Women’s Hospital, Boston, MA
| |
Collapse
|
104
|
Abstract
Hepatocellular carcinoma (HCC) is the most commonly diagnosed malignancy of the liver and is the third most frequent cause of cancer death worldwide. Although advances in HCC detection and treatment have increased the likelihood of a cure at early stages of the disease, HCC remains largely incurable because of late presentation and tumor recurrence. Only 25% of HCC patients are deemed suitable for curative treatment, with the overall survival at just a few months for inoperable patients. Additionally, this disease is particularly difficult to treat because of the high recurrence rate, its chemotherapy-resistant nature and the premalignant nature of surrounding cirrhotic liver disease. In the past few years, compelling evidence has emerged in support of the hierarchic cancer stem cell (CSC)/tumor-initiating cell (T-IC) model for solid tumors, including HCC. Understanding the characteristics and function of CSCs in the liver has also shed light on HCC management and treatment, including the implications for prognosis, prediction and treatment resistance. In this review, a detailed summary of the recent progress in liver CSC research with regard to identification, regulation and therapeutic implications will be discussed.
Collapse
Affiliation(s)
- Carol Man Tong
- Department of Pathology, Faculty of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong
| | | | | |
Collapse
|
105
|
Structure, function, and mechanism of progranulin; the brain and beyond. J Mol Neurosci 2011; 45:538-48. [PMID: 21691802 DOI: 10.1007/s12031-011-9569-4] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2011] [Accepted: 05/31/2011] [Indexed: 12/13/2022]
Abstract
Mutation of human GRN, the gene encoding the secreted glycoprotein progranulin, results in a form of frontotemporal lobar degeneration that is characterized by the presence of ubiquitinated inclusions containing phosphorylated and cleaved fragments of the transactivation response element DNA-binding protein-43. This has stimulated interest in understanding the role of progranulin in the central nervous system, and in particular, how this relates to neurodegeneration. Progranulin has many roles outside the brain, including regulation of cellular proliferation, survival, and migration, in cancer, including cancers of the brain, in wound repair, and inflammation. It often acts through the extracellular signal-regulated kinase and phopshatidylinositol-3-kinases pathways. The neurobiology of progranulin has followed a similar pattern with proposed roles for progranulin (PGRN) in the central nervous system as a neuroprotective agent and in neuroinflammation. Here we review the structure, biology, and mechanism of progranulin action. By understanding PGRN in a wider context, we may be better able to delineate its roles in the normal brain and in neurodegenerative disease.
Collapse
|
106
|
Wilson BJ, Schatton T, Frank MH, Frank NY. Colorectal Cancer Stem Cells: Biology and Therapeutic Implications. CURRENT COLORECTAL CANCER REPORTS 2011; 7:128-135. [PMID: 21552371 PMCID: PMC3087297 DOI: 10.1007/s11888-011-0093-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The hypothesis that cancer is driven by a subpopulation of tumor-initiating or cancer stem cells (CSC), defined by their selective ability for extensive self-renewal and capacity to give rise to nontumorigenic cancer cell progeny through differentiation, has been validated experimentally in diverse human malignancies. Translational relevance of the CSC hypothesis is underlined by emerging novel strategies designed to target all subpopulations within a given tumor in order to effect cancer eradication and improve patient outcomes. Colorectal cancer stem cells (CRSCs) have been identified and successfully isolated by several research groups based on distinct cell-surface marker characteristics. Identification of CRSC populations has led to a wave of discoveries describing novel self-renewal and drug resistance mechanisms in colorectal cancer that represent novel future therapeutic targets. In this review, we will discuss emerging CRSC-specific pathways and the therapeutic promise of targeting this cancer population in colorectal cancer patients.
Collapse
Affiliation(s)
- Brian J Wilson
- Transplantation Research Center, Children's Hospital Boston and Harvard Skin Disease Research Center, Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, 300 Longwood Avenue, Enders 814, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
107
|
Cellular effects of progranulin in health and disease. J Mol Neurosci 2011; 45:549-60. [PMID: 21611805 DOI: 10.1007/s12031-011-9553-z] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2011] [Accepted: 05/10/2011] [Indexed: 12/12/2022]
Abstract
Progranulin is a fascinating multifunctional protein, which has been implicated in cell growth, wound repair, tumorigenesis, inflammation, neurodevelopment, and more recently in neurodegeneration. The mechanism of action of this protein is still largely unknown, but the knowledge about the cellular effects on various cell types is expanding. In the current review, we will summarize what is known about the cell biology of progranulin. A better understanding of the biology of progranulin will impact diverse areas of research.
Collapse
|
108
|
Oishi N, Wang XW. Novel therapeutic strategies for targeting liver cancer stem cells. Int J Biol Sci 2011; 7:517-35. [PMID: 21552419 PMCID: PMC3088875 DOI: 10.7150/ijbs.7.517] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2011] [Accepted: 04/14/2011] [Indexed: 12/15/2022] Open
Abstract
The cancer stem cell (CSC) hypothesis was first proposed over 40 years ago. Advances in CSC isolation were first achieved in hematological malignancies, with the first CSC demonstrated in acute myeloid leukemia. However, using similar strategies and technologies, and taking advantage of available surface markers, CSCs have been more recently demonstrated in a growing range of epithelial and other solid organ malignancies, suggesting that the majority of malignancies are dependent on such a compartment.Primary liver cancer consists predominantly of hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (ICC). It is believed that hepatic progenitor cells (HPCs) could be the origin of some HCCs and ICCs. Furthermore, stem cell activators such as Wnt/β-catenin, TGF-β, Notch and Hedgehog signaling pathways also expedite tumorigenesis, and these pathways could serve as molecular targets to assist in designing cancer prevention strategies. Recent studies indicate that additional factors such as EpCAM, Lin28 or miR-181 may also contribute to HCC progression by targeting HCC CSCs. Various therapeutic drugs that directly modulate CSCs have been examined in vivo and in vitro. However, CSCs clearly have a complex pathogenesis, with a considerable crosstalk and redundancy in signaling pathways, and hence targeting single molecules or pathways may have a limited benefit for treatment. Many of the key signaling molecules are shared by both CSCs and normal stem cells, which add further challenges for designing molecularly targeted strategies specific to CSCs but sparing normal stem cells to avoid side effects. In addition to the direct control of CSCs, many other factors that are needed for the maintenance of CSCs, such as angiogenesis, vasculogenesis, invasion and migration, hypoxia, immune evasion, multiple drug resistance, and radioresistance, should be taken into consideration when designing therapeutic strategies for HCC. Here we provide a brief review of molecular signaling in liver CSCs and present insights into new therapeutic strategies for targeting liver CSCs.
Collapse
Affiliation(s)
| | - Xin Wei Wang
- Liver Carcinogenesis Section, Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
109
|
Frank NY, Schatton T, Kim S, Zhan Q, Wilson BJ, Ma J, Saab KR, Osherov V, Widlund HR, Gasser M, Waaga-Gasser AM, Kupper TS, Murphy GF, Frank MH. VEGFR-1 expressed by malignant melanoma-initiating cells is required for tumor growth. Cancer Res 2011; 71:1474-85. [PMID: 21212411 PMCID: PMC3083845 DOI: 10.1158/0008-5472.can-10-1660] [Citation(s) in RCA: 129] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Melanoma growth is driven by malignant melanoma-initiating cells (MMIC) identified by expression of the ATP-binding cassette (ABC) member ABCB5. ABCB5(+) melanoma subpopulations have been shown to overexpress the vasculogenic differentiation markers CD144 (VE-cadherin) and TIE1 and are associated with CD31(-) vasculogenic mimicry (VM), an established biomarker associated with increased patient mortality. Here we identify a critical role for VEGFR-1 signaling in ABCB5(+) MMIC-dependent VM and tumor growth. Global gene expression analyses, validated by mRNA and protein determinations, revealed preferential expression of VEGFR-1 on ABCB5(+) tumor cells purified from clinical melanomas and established melanoma lines. In vitro, VEGF induced the expression of CD144 in ABCB5(+) subpopulations that constitutively expressed VEGFR-1 but not in ABCB5(-) bulk populations that were predominantly VEGFR-1(-). In vivo, melanoma-specific shRNA-mediated knockdown of VEGFR-1 blocked the development of ABCB5(+) VM morphology and inhibited ABCB5(+) VM-associated production of the secreted melanoma mitogen laminin. Moreover, melanoma-specific VEGFR-1 knockdown markedly inhibited tumor growth (by > 90%). Our results show that VEGFR-1 function in MMIC regulates VM and associated laminin production and show that this function represents one mechanism through which MMICs promote tumor growth.
Collapse
Affiliation(s)
- Natasha Y. Frank
- Division of Genetics, Brigham and Women’s Hospital, Boston, Massachusetts
- Department of Medicine, Boston VA Healthcare System, Boston, Massachusetts
- Transplantation Research Center, Children’s Hospital Boston, Boston, Massachusetts
| | - Tobias Schatton
- Transplantation Research Center, Children’s Hospital Boston, Boston, Massachusetts
- Department of Dermatology, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Soo Kim
- Transplantation Research Center, Children’s Hospital Boston, Boston, Massachusetts
| | - Qian Zhan
- Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Brian J. Wilson
- Transplantation Research Center, Children’s Hospital Boston, Boston, Massachusetts
- Department of Dermatology, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Jie Ma
- Transplantation Research Center, Children’s Hospital Boston, Boston, Massachusetts
- Department of Dermatology, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Karim R. Saab
- Transplantation Research Center, Children’s Hospital Boston, Boston, Massachusetts
- Department of Dermatology, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Veronika Osherov
- Transplantation Research Center, Children’s Hospital Boston, Boston, Massachusetts
| | - Hans R. Widlund
- Department of Dermatology, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Martin Gasser
- Department of Surgery, University of Würzburg, Würzburg, Germany
| | | | - Thomas S. Kupper
- Department of Dermatology, Brigham and Women’s Hospital, Boston, Massachusetts
| | - George F. Murphy
- Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Markus H. Frank
- Transplantation Research Center, Children’s Hospital Boston, Boston, Massachusetts
- Department of Dermatology, Brigham and Women’s Hospital, Boston, Massachusetts
| |
Collapse
|
110
|
Moitra K, Scally M, McGee K, Lancaster G, Gold B, Dean M. Molecular evolutionary analysis of ABCB5: the ancestral gene is a full transporter with potentially deleterious single nucleotide polymorphisms. PLoS One 2011; 6:e16318. [PMID: 21298007 PMCID: PMC3029322 DOI: 10.1371/journal.pone.0016318] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2010] [Accepted: 12/17/2010] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND ABCB5 is a member of the ABC protein superfamily, which includes the transporters ABCB1, ABCC1 and ABCG2 responsible for causing drug resistance in cancer patients and also several other transporters that have been linked to human disease. The ABCB5 full transporter (ABCB5.ts) is expressed in human testis and its functional significance is presently unknown. Another variant of this transporter, ABCB5 beta possess a "half-transporter-like" structure and is expressed in melanoma stem cells, normal melanocytes, and other types of pigment cells. ABCB5 beta has important clinical implications, as it may be involved with multidrug resistance in melanoma stem cells, allowing these stem cells to survive chemotherapeutic regimes. METHODOLOGY/PRINCIPAL FINDINGS We constructed and examined in detail topological structures of the human ABCB5 protein and determined in-silico the cSNPs (coding single nucleotide polymorphisms) that may affect its function. Evolutionary analysis of ABCB5 indicated that ABCB5, ABCB1, ABCB4, and ABCB11 share a common ancestor, which began duplicating early in the evolutionary history of chordates. This suggests that ABCB5 has evolved as a full transporter throughout its evolutionary history. CONCLUSIONS/SIGNIFICANCE From our in-silco analysis of cSNPs we found that a large number of non-synonymous cSNPs map to important functional regions of the protein suggesting that these SNPs if present in human populations may play a role in diseases associated with ABCB5. From phylogenetic analyses, we have shown that ABCB5 evolved as a full transporter throughout its evolutionary history with an absence of any major shifts in selection between the various lineages suggesting that the function of ABCB5 has been maintained during mammalian evolution. This finding would suggest that ABCB5 beta may have evolved to play a specific role in human pigment cells and/or melanoma cells where it is predominantly expressed.
Collapse
Affiliation(s)
- Karobi Moitra
- Laboratory of Experimental Immunology, Human Genetics Section, Cancer and Inflammation Program, National Cancer Institute at Frederick, Frederick, Maryland, United States of America
| | - Mark Scally
- Laboratory of Experimental Immunology, Human Genetics Section, Cancer and Inflammation Program, National Cancer Institute at Frederick, Frederick, Maryland, United States of America
| | - Kate McGee
- Laboratory of Experimental Immunology, Human Genetics Section, Cancer and Inflammation Program, National Cancer Institute at Frederick, Frederick, Maryland, United States of America
| | - Germaine Lancaster
- Laboratory of Experimental Immunology, Human Genetics Section, Cancer and Inflammation Program, National Cancer Institute at Frederick, Frederick, Maryland, United States of America
| | - Bert Gold
- Laboratory of Experimental Immunology, Human Genetics Section, Cancer and Inflammation Program, National Cancer Institute at Frederick, Frederick, Maryland, United States of America
| | - Michael Dean
- Laboratory of Experimental Immunology, Human Genetics Section, Cancer and Inflammation Program, National Cancer Institute at Frederick, Frederick, Maryland, United States of America
- * E-mail:
| |
Collapse
|
111
|
Ma J, Lin JY, Alloo A, Wilson BJ, Schatton T, Zhan Q, Murphy GF, Waaga-Gasser AM, Gasser M, Hodi FS, Frank NY, Frank MH. Isolation of tumorigenic circulating melanoma cells. Biochem Biophys Res Commun 2010; 402:711-7. [PMID: 20977885 PMCID: PMC2998991 DOI: 10.1016/j.bbrc.2010.10.091] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Accepted: 10/19/2010] [Indexed: 12/19/2022]
Abstract
Circulating tumor cells (CTC) have been identified in several human malignancies, including malignant melanoma. However, whether melanoma CTC are tumorigenic and cause metastatic progression is currently unknown. Here, we isolate for the first time viable tumorigenic melanoma CTC and demonstrate that this cell population is capable of metastasis formation in human-to-mouse xenotransplantation experiments. The presence of CTC among peripheral blood mononuclear cells (PBMC) of murine recipients of subcutaneous (s.c.) human melanoma xenografts could be detected based on mRNA expression for human GAPDH and/or ATP-binding cassette subfamily B member 5 (ABCB5), a marker of malignant melanoma-initiating cells previously shown to be associated with metastatic disease progression in human patients. ABCB5 expression could also be detected in PBMC preparations from human stage IV melanoma patients but not healthy controls. The detection of melanoma CTC in human-to-mouse s.c. tumor xenotransplantation models correlated significantly with pulmonary metastasis formation. Moreover, prospectively isolated CTC from murine recipients of s.c. melanoma xenografts were capable of primary tumor initiation and caused metastasis formation upon xenotransplantation to secondary murine NOD-scid IL2Rγ(null) recipients. Our results provide initial evidence that melanoma CTC are tumorigenic and demonstrate that CTC are capable of causing metastatic tumor progression. These findings suggest a need for CTC eradication to inhibit metastatic progression and provide a rationale for assessment of therapeutic responses of this tumorigenic cell population to promising emerging melanoma treatment modalities.
Collapse
Affiliation(s)
- Jie Ma
- Transplantation Research Center, Children’s Hospital Boston, Harvard Medical School, Boston, MA, USA
- Department of Dermatology, Brigham & Women’s Hospital, Boston, MA, USA
| | - Jennifer Y. Lin
- Department of Dermatology, Brigham & Women’s Hospital, Boston, MA, USA
| | - Allireza Alloo
- Transplantation Research Center, Children’s Hospital Boston, Harvard Medical School, Boston, MA, USA
- Department of Dermatology, Brigham & Women’s Hospital, Boston, MA, USA
| | - Brian J. Wilson
- Transplantation Research Center, Children’s Hospital Boston, Harvard Medical School, Boston, MA, USA
- Department of Dermatology, Brigham & Women’s Hospital, Boston, MA, USA
| | - Tobias Schatton
- Transplantation Research Center, Children’s Hospital Boston, Harvard Medical School, Boston, MA, USA
- Department of Dermatology, Brigham & Women’s Hospital, Boston, MA, USA
| | - Qian Zhan
- Department of Pathology, Brigham & Women’s Hospital, Boston, MA, USA
| | - George F. Murphy
- Department of Pathology, Brigham & Women’s Hospital, Boston, MA, USA
| | | | - Martin Gasser
- Department of Surgery, University of Würzburg Medical School, Würzburg, Germany
| | - F. Stephen Hodi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Natasha Y. Frank
- Department of Medicine, VA Boston Healthcare System, Boston, MA, USA
- Division of Genetics, Brigham & Women’s Hospital, Boston, MA, USA
| | - Markus H. Frank
- Transplantation Research Center, Children’s Hospital Boston, Harvard Medical School, Boston, MA, USA
- Department of Dermatology, Brigham & Women’s Hospital, Boston, MA, USA
| |
Collapse
|
112
|
Pearce-McCall D, Newman JP. Expectation of success following noncontingent punishment in introverts and extraverts. J Pers Soc Psychol 1986; 2:17. [PMID: 23815814 PMCID: PMC3701589 DOI: 10.1186/2162-3619-2-17] [Citation(s) in RCA: 228] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Accepted: 06/25/2013] [Indexed: 12/14/2022]
Abstract
Recent findings indicate that extraverts are more likely than introverts to continue responding in the face of punishment and frustrating nonreward (Newman & Kosson, 1984; Tiggemann, Winefield, & Brebner, 1982). The current study investigates whether extraverts' expectations for success are, similarly, resistant to interruption and alteration. To test this hypothesis, 50 introverted and 50 extraverted male undergraduates were exposed to pretreatment with either a 50% level of noncontingent reward or a 50% level of noncontingent punishment. As predicted, there were significant Group X Pretreatment interactions on all dependent measures. In comparison to those introverts who received the punishment pretreatment, extraverts exposed to the same pretreatment placed larger wagers on their ability to succeed, and reported higher levels of perceived control. In addition, relative to their estimates for the pretreatment task, extraverts exposed to noncontingent punishment increased their expectation for success, whereas introverts exposed to noncontingent punishment decreased their performance expectations. No differences were observed between the two groups following pretreatment with noncontingent reward. The results suggest that extraverts are characterized by a distinctive reaction to punishment involving response facilitation as opposed to response inhibition.
Collapse
|