Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Am J Pathol ; 191(2): 335-352, 2021 02.
Article in English | MEDLINE | ID: mdl-33181139

ABSTRACT

Human T-lymphotropic virus type 1 (HTLV-1) causes adult T-cell leukemia, a disease commonly associated with hypercalcemia and osteolysis. There is no effective treatment for HTLV-1, and the osteolytic mechanisms are not fully understood. Mice expressing the HTLV-1 oncogene Tax, driven by the human granzyme B promoter (Tax+), develop osteolytic tumors. To investigate the progression of the bone-invasive malignancies, wild-type, Tax+, and Tax+/interferon-γ-/- mice were assessed using necropsy, histologic examination, IHC analysis, flow cytometry, and advanced imaging. Tax+ and Tax+/interferon-γ-/- malignancies of the ear, tail, and foot comprised poorly differentiated, round to spindle-shaped cells with prominent neutrophilic infiltrates. Tail tumors originated from muscle, nerve, and/or tendon sheaths, with frequent invasion into adjacent bone. F4/80+ and anti-mouse CD11b (Mac-1)+ histiocytic cells predominated within the tumors. Three Tax+/interferon-γ-/- cell lines were generated for in vivo allografts, in vitro gene expression and bone resorption assays. Two cell lines were of monocyte/macrophage origin, and tumors formed in vivo in all three. Differences in Pthrp, Il6, Il1a, Il1b, and Csf3 expression in vitro were correlated with differences in in vivo plasma calcium levels, tumor growth, metastasis, and neutrophilic inflammation. Tax+ mouse tumors were classified as bone-invasive histiocytic sarcomas. The cell lines are ideal for further examination of the role of HTLV-1 Tax in osteolytic tumor formation and the development of hypercalcemia and tumor-associated inflammation.


Subject(s)
Cell Line, Tumor , Disease Models, Animal , Genes, pX , HTLV-I Infections/complications , Histiocytic Sarcoma , Animals , Carcinogenesis/genetics , Histiocytic Sarcoma/pathology , Histiocytic Sarcoma/virology , Human T-lymphotropic virus 1/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oncogenes , Osteolysis/pathology , Osteolysis/virology
2.
Article in English | MEDLINE | ID: mdl-26893580

ABSTRACT

INTRODUCTION: Interleukin-6 (IL-6) is an important growth factor for estrogen receptor-α (ERα)-positive breast cancer, and elevated serum IL-6 is associated with poor prognosis. METHODS: The role of the phosphorylated signal transducer and activator of transcription 3 pathway was investigated in ERα-positive breast cancer. A panel of cell lines was treated with exogenous IL-6. An IL-6 specific gene signature was generated by profiling ten ERα-positive breast cancer cell lines alone or following treatment with 10 ng/mL recombinant IL-6 or human marrow stromal cell-conditioned media, with or without siltuximab (a neutralizing anti-IL-6 antibody) and grown in three-dimensional tumor microenvironment-aligned cultures for 4 days, 5 days, or 6 days. The established IL-6 signature was validated against 36 human ERα-positive breast tumor samples with matched serum. A comparative MCF-7 xenograft murine model was utilized to determine the role of IL-6 in estrogen-supplemented ERα-positive breast cancer to assess the efficacy of anti-IL-6 therapy in vivo. RESULTS: In eight of nine ERα-positive breast cancer cell lines, recombinant IL-6 increased phosphorylation of tyrosine 705 of STAT3. Differential gene expression analysis identified 17 genes that could be used to determine IL-6 pathway activation by combining their expression intensity into a pathway activation score. The gene signature included a variety of genes involved in immune cell function and migration, cell growth and apoptosis, and the tumor microenvironment. Validation of the IL-6 gene signature in 36 matched human serum and ERα-positive breast tumor samples showed that patients with a high IL-6 pathway activation score were also enriched for elevated serum IL-6 (≥10 pg/mL). When human IL-6 was provided in vivo, MCF-7 cells engrafted without the need for estrogen supplementation, and addition of estrogen to IL-6 did not further enhance engraftment. Subsequently, we prophylactically treated mice at MCF-7 engraftment with siltuximab, fulvestrant, or combination therapy. Siltuximab alone was able to blunt MCF-7 engraftment. Similarly, siltuximab alone induced regressions in 90% (9/10) of tumors, which were established in the presence which were established in the presence of hMSC expressing human IL-6 and estrogen. CONCLUSION: Given the established role for IL-6 in ERα-positive breast cancer, these data demonstrate the potential for anti-IL-6 therapeutics in breast cancer.

3.
Cell Stem Cell ; 14(3): 357-69, 2014 Mar 06.
Article in English | MEDLINE | ID: mdl-24607407

ABSTRACT

Stem-like glioma cells reside within a perivascular niche and display hallmark radiation resistance. An understanding of the mechanisms underlying these properties will be vital for the development of effective therapies. Here, we show that the stem cell marker CD44 promotes cancer stem cell phenotypes and radiation resistance. In a mouse model of glioma, Cd44(-/-) and Cd44(+/-) animals showed improved survival compared to controls. The CD44 ligand osteopontin shared a perivascular expression pattern with CD44 and promoted glioma stem cell-like phenotypes. These effects were mediated via the γ-secretase-regulated intracellular domain of CD44, which promoted aggressive glioma growth in vivo and stem cell-like phenotypes via CBP/p300-dependent enhancement of HIF-2α activity. In human glioblastoma multiforme, expression of CD44 correlated with hypoxia-induced gene signatures and poor survival. Altogether, these data suggest that in the glioma perivascular niche, osteopontin promotes stem cell-like properties and radiation resistance in adjacent tumor cells via activation of CD44 signaling.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , Hyaluronan Receptors/metabolism , Neoplastic Stem Cells/pathology , Osteopontin/metabolism , Signal Transduction , Stem Cell Niche , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , E1A-Associated p300 Protein/metabolism , Glioblastoma/metabolism , Humans , Hyaluronan Receptors/chemistry , Ligands , Mice , Neoplasm Invasiveness , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Phenotype , Platelet-Derived Growth Factor/pharmacology , Protein Structure, Tertiary , Signal Transduction/drug effects , Stem Cell Niche/drug effects , Survival Analysis
4.
Oral Oncol ; 48(6): 491-9, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22265717

ABSTRACT

Oral squamous cell carcinoma (OSCC) is the most commonly diagnosed oral malignancy in humans and cats and frequently invades bone. The objective of this study was to determine if feline OSCC serves as a relevant model of human OSCC in terms of osteolytic behavior and expression of bone resorption agonists. Novel feline OSCC cell lines (SCCF2 and SCCF3) were derived from spontaneous carcinomas. Gene expression and osteolytic behavior were compared to an established feline OSCC cell line (SCCF1) and three human OSCC cell lines (UMSCC-12, A253 and SCC25). Interaction of OSCC with bone and murine pre-osteoblasts (MC3T3) was investigated using in vitro co-culture techniques. In vivo bioluminescent imaging, Faxitron radiography and microscopy were used to measure xenograft growth and bone invasion in nude mice. Human and feline OSCC expressing the highest levels of parathyroid hormone-related protein (PTHrP) were associated with in vitro and in vivo bone resorption and osteoclastogenesis. MC3T3 cells had increased receptor activator of nuclear factor κB ligand (RANKL) expression and reduced osteoprotegerin (OPG) expression in conditioned medium from bone-invasive SCCF2 cells compared to minimally bone invasive SCCF3 cells, which was partially reversed with a neutralizing anti-PTHrP antibody. Human and feline OSCC cells cultured in bone-conditioned medium had increased PTHrP secretion and proliferation. Feline OSCC-induced bone resorption was associated with tumor cell secretion of PTHrP and with increased RANKL:OPG expression ratio in mouse preosteoblasts. Bone-CM increased OSCC proliferation and secretion of PTHrP. The preclinical models of feline OSCC recapitulated the bone-invasive phenotype characteristic of spontaneous OSCC and will be useful to future preclinical and mechanistic studies of bone invasive behavior.


Subject(s)
Bone Resorption/metabolism , Carcinoma, Squamous Cell/metabolism , Mouth Neoplasms/metabolism , Animals , Bone Resorption/veterinary , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/veterinary , Cats , Cell Line, Tumor , Disease Models, Animal , Humans , Mice , Mouth Neoplasms/pathology , Mouth Neoplasms/veterinary , Osteoprotegerin/metabolism , Parathyroid Hormone-Related Protein/metabolism , RANK Ligand/metabolism
5.
Leuk Lymphoma ; 53(4): 688-98, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21942940

ABSTRACT

Parathyroid hormone-related protein (PTHrP) and macrophage inflammatory protein-1α (MIP-1α) have been implicated in the pathogenesis of adult T-cell leukemia/lymphoma, but their effects on T-cells have not been well studied. Here we analyzed the functions of PTHrP and MIP-1α on T-cell growth and death both in vitro and in vivo by overexpressing either factor in human Jurkat T-cells. PTHrP or MIP-1α did not affect Jurkat cell growth in vitro, but PTHrP increased their sensitivity to apoptosis. Importantly, PTHrP and MIP-1α decreased both tumor incidence and growth in vivo. To investigate possible mechanisms, polymerase chain reaction (PCR) arrays and real-time reverse transcription (RT)-PCR assays were performed. Both PTHrP and MIP-1α increased the expression of several factors including signal transducer and activator of transcription 4, tumor necrosis factor α, receptor activator of nuclear factor κB ligand and death-associated protein kinase 1, and decreased the expression of inhibitor of DNA binding 1, interferon γ and CD40 ligand in Jurkat cells. In addition, MIP-1α also increased the expression of transcription factor AP-2α and PTHrP increased expression of the vitamin D3 receptor. These data demonstrate that PTHrP and MIP-1α exert a profound antitumor effect presumably by increasing the sensitivity to apoptotic signals through modulation of transcription and apoptosis factors in T-cells.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Chemokine CCL3/genetics , Gene Expression Regulation, Leukemic , Leukemia, Experimental/genetics , Parathyroid Hormone-Related Protein/genetics , Animals , Apoptosis/genetics , CD40 Ligand/genetics , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Cell Line, Tumor , Death-Associated Protein Kinases , Humans , Interferon-gamma/genetics , Jurkat Cells , Leukemia, Experimental/pathology , Leukemia, T-Cell/genetics , Leukemia, T-Cell/pathology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Oligonucleotide Array Sequence Analysis , RANK Ligand/genetics , Reverse Transcriptase Polymerase Chain Reaction , STAT4 Transcription Factor/genetics , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Transplantation, Heterologous , Tumor Necrosis Factor-alpha/genetics
6.
Clin Exp Metastasis ; 28(4): 377-89, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21374084

ABSTRACT

Osteosarcoma (OSA) is an aggressive, highly metastatic and lytic primary bone neoplasm commonly affecting the appendicular skeleton of dogs and children. Current treatment options include amputation of the afflicted limb, limb-sparing procedures, or palliative radiation with or without adjunct chemotherapy. Therapies that inhibit bone resorption, such as the bisphosphonates, may be an effective palliative therapy by limiting the local progression of OSA in those patients that are not viable candidates for amputation. We have developed a mouse model of canine skeletal OSA following intratibial inoculation of OSCA40 cells that spontaneously metastasized to the lungs. We demonstrated that therapy with a nitrogen-containing bisphosphonate, zoledronic acid (Zol), reduced OSA-induced bone lysis; however, Zol monotherapy or in combination with amputation was not effective at inhibiting pulmonary metastasis. While not reaching statistical significance, amputation of the tumor-bearing limb reduced the average incidence of lung metastases; however, this effect was nullified when Zol was added to the treatment protocol. In untreated mice, the magnitude of proximal tibial lysis was significantly correlated with the incidence of metastasis. The data support amputation alone for the management of appendicular OSA rather than combining amputation with Zol. However, in patients that are not viable candidates for amputation, Zol may be a useful palliative therapy for OSA by reducing the magnitude of lysis and therefore bone pain, despite the risk of increased pulmonary metastasis.


Subject(s)
Amputation, Surgical , Antineoplastic Agents/pharmacology , Bone Neoplasms/drug therapy , Bone Neoplasms/surgery , Diphosphonates/pharmacology , Dog Diseases , Imidazoles/pharmacology , Lung Neoplasms/secondary , Osteosarcoma/veterinary , Animals , Bone Neoplasms/pathology , Bone Neoplasms/veterinary , Dog Diseases/drug therapy , Dog Diseases/pathology , Dog Diseases/surgery , Dogs , Female , Lung Neoplasms/therapy , Mice , Mice, Nude , Neoplasm Invasiveness/pathology , Osteosarcoma/drug therapy , Osteosarcoma/pathology , Osteosarcoma/surgery , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Zoledronic Acid
7.
Prostate ; 71(12): 1251-63, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21321976

ABSTRACT

BACKGROUND: Prostate cancer in men has a high mortality and morbidity due to metastatic disease. The pathobiology of prostate cancer metastasis is not well understood and cell lines and animal models that recapitulate the complex nature of the disease are needed. Therefore, the goal of the study was to establish and characterize a new prostate cancer line derived from a dog with spontaneous prostate cancer. METHODS: A new cell line (Leo) was derived from a dog with spontaneous prostate cancer. Immunohistochemistry and PCR were used to characterize the primary prostate cancer and xenografts in nude mice. Subcutaneous tumor growth and metastases in nude mice were evaluated by bioluminescent imaging, radiography and histopathology. In vitro chemosensitivity of Leo cells to therapeutic agents was measured. RESULTS: Leo cells expressed the secretory epithelial cytokeratins (CK)8, 18, and ductal cell marker, CK7. The cell line grew in vitro (over 75 passages) and was tumorigenic in the subcutis of nude mice. Following intracardiac injection, Leo cells metastasized to the brain, spinal cord, bone, and adrenal gland. The incidence of metastases was greatest to the central nervous system (80%) with a lower incidence to bone (20%) and the adrenal glands (16%). In vitro chemosensitivity assays demonstrated that Leo cells were sensitive to Velcade and an HDAC-42 inhibitor with IC(50) concentrations of 1.9 nm and 0.95 µm, respectively. CONCLUSION: The new prostate cancer cell line (Leo) will be a valuable model to investigate the mechanisms of the brain and bone metastases.


Subject(s)
Brain Neoplasms/pathology , Brain Neoplasms/secondary , Carcinoma/pathology , Carcinoma/secondary , Cell Line, Tumor , Prostatic Neoplasms/pathology , Adrenal Gland Neoplasms/epidemiology , Adrenal Gland Neoplasms/secondary , Animals , Antineoplastic Agents/pharmacology , Bone Neoplasms/epidemiology , Bone Neoplasms/secondary , Boronic Acids/pharmacology , Bortezomib , Brain Neoplasms/epidemiology , Brain Neoplasms/metabolism , Carcinogenicity Tests , Carcinoma/epidemiology , Carcinoma/metabolism , Cell Division , Cell Line, Tumor/drug effects , Cell Line, Tumor/pathology , Dogs , Immunohistochemistry , Incidence , Injections, Subcutaneous , Keratin-18/metabolism , Keratin-7/metabolism , Keratin-8/metabolism , Male , Mice , Mice, Nude , Neoplasm Transplantation , Neoplasms, Connective Tissue/secondary , Parathyroid Hormone-Related Protein/metabolism , Phenylbutyrates/antagonists & inhibitors , Pyrazines/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Spinal Cord Neoplasms/epidemiology , Spinal Cord Neoplasms/secondary , Subcutaneous Tissue , Transplantation, Heterologous
8.
Prostate ; 71(6): 615-25, 2011 May.
Article in English | MEDLINE | ID: mdl-20957670

ABSTRACT

BACKGROUND: Osteoblastic bone metastasis is the predominant phenotype observed in prostate cancer patients and is associated with high patient mortality and morbidity. However, the mechanisms determining the development of this phenotype are not well understood. Prostate cancer cells secrete several osteogenic factors including Wnt proteins, which are not only osteoinductive but also oncogenic. Therefore, the purpose of the study was to investigate the contribution of the Wnt signaling pathway in prostate cancer growth, incidence of bone metastases, and osteoblastic phenotype of bone metastases. The strategy involved overexpressing the Wnt antagonist, DKK-1, in the mixed osteoblastic and osteolytic Ace-1 prostate cancer cells. METHODS: Ace-1 prostate cancer cells stably expressing human DKK-1 or empty vector were established and transduced with lentiviral yellow fluorescent protein (YFP)-luciferase (Luc). The Ace-1/vector(YFP-LUC) and Ace-1/DKK-1(YFP-LUC) cells were injected subcutaneously, intratibially, or in the left cardiac ventricle in athymic mice. RESULTS: Unexpectedly, DKK-1 significantly increased Ace-1 subcutaneous tumor mass and the incidence of bone metastases after intracardiac injection of Ace-1 cells. DKK-1 increased Ace-1 tumor growth associated with increased phospho46 c-Jun amino-terminal kinase by the Wnt noncanonical pathway. As expected, DKK-1 decreased the Ace-1 osteoblastic phenotype of bone metastases, as confirmed by radiographic, histopathologic, and microcomputed tomographic analysis. DKK-1 decreased osteoblastic activity via the Wnt canonical pathway evidenced by an inhibition of T-cell factor activity in murine osteoblast precursor ST2 cells. CONCLUSION: The present study showed that DKK-1 is a potent inhibitor of bone growth in prostate cancer-induced osteoblastic metastases.


Subject(s)
Bone Neoplasms/secondary , Intercellular Signaling Peptides and Proteins/metabolism , Prostatic Neoplasms/pathology , Wnt Proteins/metabolism , Animals , Bone Neoplasms/metabolism , Cell Growth Processes/physiology , Cell Line, Tumor , Dogs , Histocytochemistry , Intercellular Signaling Peptides and Proteins/genetics , Male , Mice , Mice, Nude , Osteogenesis/physiology , Prostatic Neoplasms/metabolism , Signal Transduction , Statistics, Nonparametric , Tomography, X-Ray Computed , Wnt Proteins/antagonists & inhibitors
9.
Cancer Res ; 70(21): 8607-16, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20959474

ABSTRACT

Squamous cell carcinoma (SCC) is the most common form of oral cancer. Destruction and invasion of mandibular and maxillary bone frequently occurs and contributes to morbidity and mortality. We hypothesized that the bisphosphonate drug zoledronic acid (ZOL) would inhibit tumor-induced osteolysis and reduce tumor growth and invasion in a murine xenograft model of bone-invasive oral SCC (OSCC) derived from an osteolytic feline OSCC. Luciferase-expressing OSCC cells (SCCF2Luc) were injected into the perimaxillary subgingiva of nude mice, which were then treated with 100 µg/kg ZOL or vehicle. ZOL treatment reduced tumor growth and prevented loss of bone volume and surface area but had no effect on tumor invasion. Effects on bone were associated with reduced osteolysis and increased periosteal new bone formation. ZOL-mediated inhibition of tumor-induced osteolysis was characterized by reduced numbers of tartrate-resistant acid phosphatase-positive osteoclasts at the tumor-bone interface, where it was associated with osteoclast vacuolar degeneration. The ratio of eroded to total bone surface was not affected by treatment, arguing that ZOL-mediated inhibition of osteolysis was independent of effects on osteoclast activation or initiation of bone resorption. In summary, our results establish that ZOL can reduce OSCC-induced osteolysis and may be valuable as an adjuvant therapy in OSCC to preserve mandibular and maxillary bone volume and function.


Subject(s)
Bone Resorption/prevention & control , Carcinoma, Squamous Cell/prevention & control , Diphosphonates/therapeutic use , Disease Models, Animal , Imidazoles/therapeutic use , Mouth Neoplasms/prevention & control , Osteolysis/prevention & control , Acid Phosphatase , Animals , Bone Density Conservation Agents/therapeutic use , Bone Resorption/metabolism , Bone Resorption/pathology , Calcium/metabolism , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cats , Isoenzymes , Luciferases/metabolism , Male , Mice , Mice, Nude , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Osteoclasts/cytology , Osteoclasts/drug effects , Osteolysis/metabolism , Osteolysis/pathology , Tartrate-Resistant Acid Phosphatase , Transplantation, Heterologous , X-Ray Microtomography , Zoledronic Acid
10.
J Surg Res ; 162(2): e9-17, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20538301

ABSTRACT

BACKGROUND: Parathyroid hormone-related protein (PTHrP) has anabolic effects in bone, which has led to the clinical use of N-terminal fragments of PTHrP and PTH. Since 10% to 20% of fractures demonstrate healing complications and osteoporosis continues to be a debilitating disease, the development of bone-forming agents is of utmost importance. Due to evidence that regions of PTHrP other than the N-terminus may have bone-forming effects, this study was designed to compare the effects of full-length PTHrP 1-141 to N-terminal PTHrP 1-86 on in vitro bone formation. MATERIALS AND METHODS: MC3T3-E1 pre-osteoblasts were treated once every 6 d for 36 d with 5, 25, and 50 pM of PTHrP 1-141 or 1-86 for 1 or 24 h. Cells were also treated after blocking the N-terminus, the nuclear localization sequence (NLS), and the C-terminus of PTHrP, individually and in combination. Area of mineralization, alkaline phosphatase (ALP), and osteocalcin (OCN) were measured. RESULTS: PTHrP 1-141 and 1-86 increased mineralization after 24-h treatments, but not 1-h. PTHrP 1-141 was more potent than 1-86. Treatment with PTHrP 1-141 for 24-h, but not 1-86, resulted in a concentration-dependent increase in ALP, with no effect after 1-h. Exposure to both peptides for 1- or 24-h induced a concentration-dependent increase in OCN, with 24-h exceeding 1-h. Antibody blocking revealed that the NLS and C-terminus are anabolic. CONCLUSIONS: Both PTHrP 1-141 and 1-86 increased in vitro bone formation; however, PTHrP 1-141 was more effective. The NLS and C-terminus have anabolic effects distinct from the N-terminus. This demonstrates the advantage of PTHrP 1-141 as a skeletal anabolic agent.


Subject(s)
Bone Development/drug effects , Osteoblasts/physiology , Parathyroid Hormone-Related Protein/therapeutic use , Parathyroid Hormone/therapeutic use , Peptide Fragments/therapeutic use , 3T3 Cells/cytology , 3T3 Cells/drug effects , 3T3 Cells/physiology , Alkaline Phosphatase/drug effects , Alkaline Phosphatase/metabolism , Animals , Calcification, Physiologic/drug effects , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor , Cloning, Molecular , Drug Stability , Humans , Mice , Osteoblasts/cytology , Osteoblasts/drug effects , Osteocalcin/drug effects , Osteocalcin/metabolism , Parathyroid Hormone-Related Protein/genetics , Peptide Fragments/genetics , RNA, Messenger/genetics
11.
FASEB J ; 24(6): 1947-57, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20145205

ABSTRACT

The functions of parathyroid hormone-related protein (PTHrP) on morphogenesis, cell proliferation, apoptosis, and calcium homeostasis have been attributed to its N terminus. Evidence suggests that many of these effects are not mediated by the N terminus but by the midregion, a nuclear localization sequence (NLS), and C terminus of the protein. A knock-in mouse lacking the midregion, NLS, and C terminus of PTHrP (Pthrp(Delta/Delta)) was developed. Pthrp(Delta/Delta) mice had craniofacial dysplasia, chondrodysplasia, and kyphosis, with most mice dying by d 5 of age. In bone, there were fewer chondrocytes and osteoblasts per area, bone mass was decreased, and the marrow was less cellular, with erythroid hypoplasia. Cellular proliferation was impaired, and apoptosis was increased. Runx2, Ocn, Sox9, Crtl1, beta-catenin, Runx1, ephrin B2, cyclin D1, and Gata1 were underexpressed while P16/Ink4a, P21, GSK-3beta, Il-6, Ffg3, and Ihh were overexpressed. Mammary gland development was aberrant, and energy metabolism was deregulated. These results establish that the midregion, NLS, and C terminus of PTHrP are crucial for the commitment of osteogenic and hematopoietic precursors to their lineages, and for survival, and many of the effects of PTHrP on development are not mediated by its N terminus. The down-regulation of Runx1, Runx2, and Sox9 indicates that PTHrP is a modulator of transcriptional activation during stem cell commitment.


Subject(s)
Apoptosis , Bone and Bones/cytology , Genes, Lethal , Hematopoiesis , Nuclear Localization Signals/deficiency , Parathyroid Hormone-Related Protein/physiology , Animals , Blotting, Western , Bone and Bones/pathology , Chondrocytes/cytology , Chondrocytes/pathology , Female , Flow Cytometry , Gene Expression Profiling , Gene Knock-In Techniques , Immunoenzyme Techniques , Male , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoblasts/cytology , Osteoblasts/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate
12.
Cancer Res ; 68(21): 9087-95, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18974155

ABSTRACT

Common sites of breast cancer metastasis include the lung, liver, and bone, and of these secondary metastatic sites, estrogen receptor alpha (ERalpha)-positive breast cancer often favors bone. Within secondary organs, cancer cells would predictably encounter tissue-specific fibroblasts or their soluble factors, yet our understanding of how tissue-specific fibroblasts directly affect cancer cell growth rates and survival remains largely unknown. Therefore, we tested the hypothesis that mesenchymal fibroblasts isolated from common sites of breast cancer metastasis provide a more favorable microenvironment with respect to tumor growth rates. We found a direct correlation between the ability of breast, lung, and bone fibroblasts to enhance ERalpha-positive breast cancer cell growth and the level of soluble interleukin-6 (IL-6) produced by each organ-specific fibroblast, and fibroblast-mediated growth enhancement was inhibited by the removal or inhibition of IL-6. Interestingly, mice coinjected with MCF-7 breast tumor cells and senescent skin fibroblasts, which secrete IL-6, developed tumors, whereas mice coinjected with presenescent skin fibroblasts that produce little to no IL-6 failed to form xenograft tumors. We subsequently determined that IL-6 promoted growth and invasion of breast cancer cells through signal transducer and activator of transcription 3-dependent up-regulation of Notch-3, Jagged-1, and carbonic anhydrase IX. These data suggest that tissue-specific fibroblasts and the factors they produce can promote breast cancer disease progression and may represent attractive targets for development of new therapeutics.


Subject(s)
Breast Neoplasms/pathology , Cell Division/physiology , Fibroblasts/cytology , Interleukin-6/physiology , Neoplasm Invasiveness , Neoplasm Metastasis , Breast Neoplasms/metabolism , Cell Line, Tumor , Culture Media, Conditioned , Humans , Immunoprecipitation , Phosphorylation , RNA Interference , STAT3 Transcription Factor/metabolism
13.
Prostate ; 68(10): 1116-25, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18461562

ABSTRACT

BACKGROUND: Bone metastasis is the most common cause of morbidity and mortality in patients with advanced prostate cancer and is manifested primarily as mixed osteoblastic and osteolytic lesions. However, the mechanisms responsible for bone metastases in prostate cancer are not clearly understood, in part due to the lack of relevant in vivo models that mimic the clinical presentation of the disease in humans. We previously established a nude mouse model with mixed bone metastases using intracardiac injection of canine prostate cancer cells (Ace-1). In this study, we hypothesized that tumor-induced osteolysis promoted the incidence of bone metastases and osteoblastic activity. METHODS: We studied the effect of inhibition of osteolysis with zoledronic acid (ZA) on the prevention and progression of Ace-1 bone metastases in nude mice using prophylactic and delayed treatment protocols. Bioluminescent imaging, radiography, and histopathological evaluation were performed to monitor the effect of ZA on the incidence, progression and nature of bone metastases. RESULTS: Unexpectedly, there was no significant difference in tumor burden and the incidence of metastasis between control and treatment groups as detected by bioluminescent imaging and bone histomorphometry. However, radiographic and histopathological analysis showed a significant treatment-related decrease in osteolysis, but no effect on tumor-induced trabecular bone thickness in both treatment groups compared to controls. CONCLUSION: Our results demonstrated that the incidence of prostate cancer bone metastases in vivo was not reduced by zoledronic acid even though zoledronic acid inhibited bone resorption and bone loss associated with the mixed osteoblastic/osteolytic bone metastases in the Ace-1 model.


Subject(s)
Adenocarcinoma/drug therapy , Bone Density Conservation Agents/pharmacology , Bone Neoplasms/drug therapy , Diphosphonates/pharmacology , Imidazoles/pharmacology , Osteolysis/drug therapy , Prostatic Neoplasms/pathology , Adenocarcinoma/epidemiology , Adenocarcinoma/secondary , Animals , Bone Neoplasms/epidemiology , Bone Neoplasms/secondary , Cell Line, Transformed , Disease Models, Animal , Dogs , Incidence , Luminescent Proteins , Male , Mice , Mice, Nude , Osteoclasts/drug effects , Osteoclasts/pathology , Osteolysis/diagnostic imaging , Osteolysis/epidemiology , Prostatic Neoplasms/epidemiology , Radiography , Zoledronic Acid
14.
Cancer Res ; 67(24): 11859-66, 2007 Dec 15.
Article in English | MEDLINE | ID: mdl-18089816

ABSTRACT

Adult T-cell /lymphomaleukemia (ATLL) is caused by human T-cell lymphotropic virus type 1 (HTLV-1). Approximately 80% of ATLL patients develop humoral hypercalcemia of malignancy (HHM), a life-threatening complication leading to a poor prognosis. Parathyroid hormone-related protein (PTHrP) and macrophage inflammatory protein-1 alpha (MIP-1 alpha) are important factors in the pathogenesis of HHM in ATLL and the expression of PTHrP can be activated by nuclear factor kappaB (NF-kappaB). NF-kappaB is constitutively activated in ATLL cells and is essential for leukemogenesis including transformation of lymphocytes infected by HTLV-1. Our goal was to evaluate the effects of NF-kappaB disruption by a proteasomal inhibitor (PS-341) and osteoclastic inhibition by zoledronic acid (Zol) on the development of ATLL and HHM using a novel bioluminescent mouse model. We found that PS-341 decreased cell viability, increased apoptosis, and down-regulated PTHrP expression in ATLL cells in vitro. To investigate the in vivo efficacy, nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice were xenografted with ATLL cells and treated with vehicle control, PS-341, Zol, or a combination of PS-341 and Zol. Bioluminescent imaging and tumor cell count showed a significant reduction in tumor burden in mice from all treatment groups. All treatments also significantly reduced the plasma calcium concentrations. Zol treatment increased trabecular bone volume and decreased osteoclast parameters. PS-341 reduced PTHrP and MIP-1 alpha expression in tumor cells in vivo. Our results indicate that both PS-341 and Zol are effective treatments for ATLL and HHM, which are refractory to conventional therapy.


Subject(s)
HTLV-I Infections/drug therapy , HTLV-I Infections/physiopathology , Animals , Apoptosis/drug effects , Boronic Acids/therapeutic use , Bortezomib , Cell Survival/drug effects , Disease Models, Animal , Genes, Reporter , Humans , Jurkat Cells , Luciferases/genetics , Male , Mice , Mice, Inbred NOD , Mice, SCID , Protease Inhibitors/therapeutic use , Pyrazines/therapeutic use
15.
Breast J ; 9(2): 91-7, 2003.
Article in English | MEDLINE | ID: mdl-12603381

ABSTRACT

Mastalgia is a common condition that is thought to be hormonally related, but the mechanisms of pain causation are unknown. Inflammatory cytokines are implicated in pain modulation, but have not been studied with regard to mastalgia. We compared the relationship of mastalgia to the expression of the cytokines interleukin (IL)-6, IL-1beta, and tumor necrosis factor (TNF)-alpha and the degree of tissue infiltration with inflammatory cells in breast tissue from 29 premenopausal women with breast pain and 29 age-matched pain-free controls. Paraffin sections from breast biopsy samples were scored for the presence of inflammatory infiltrate and were evaluated for the expression of IL-6, IL-1beta, and TNF-alpha using standard immunohistochemical procedures. TNF-alpha and IL-6 expression displayed a trend toward slightly lower values in patients with pain (median TNF-alpha score, 3 versus 5; median IL-6 score, 3 versus 4). In the luteal phase, patients with mastalgia showed a trend toward lower expression of IL-6 (p = 0.4) in comparison to those without pain. A similar trend was also seen with TNF-alpha expression (p = 0.4). IL-1beta expression was extremely scant in the first 30 samples and was not investigated further. The degree of inflammatory infiltrate in the tissue was unrelated to the presence of breast pain. These data suggest that the three cytokines tested in this study do not play a significant role in the causation of mastalgia and lend weight to the previous finding that there are no identifiable histologic correlates of this troubling condition. Further investigation of the role of cytokines in breast pain is warranted, especially in view of the possible association between mastalgia and breast cancer risk.


Subject(s)
Breast Diseases/immunology , Breast Diseases/pathology , Interleukin-1/metabolism , Interleukin-6/metabolism , Pain/etiology , Tumor Necrosis Factor-alpha/metabolism , Adult , Breast Diseases/complications , Breast Neoplasms/etiology , Case-Control Studies , Female , Humans , Immunohistochemistry/methods , Menstrual Cycle/metabolism , Pain/immunology , Pain/pathology , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...