Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Clin Invest ; 132(12)2022 06 15.
Article in English | MEDLINE | ID: mdl-35511419

ABSTRACT

Fibrodysplasia ossificans progressiva (FOP) is a rare genetic disorder whose most debilitating pathology is progressive and cumulative heterotopic ossification (HO) of skeletal muscles, ligaments, tendons, and fascia. FOP is caused by mutations in the type I BMP receptor gene ACVR1, which enable ACVR1 to utilize its natural antagonist, activin A, as an agonistic ligand. The physiological relevance of this property is underscored by the fact that HO in FOP is exquisitely dependent on activation of FOP-mutant ACVR1 by activin A, an effect countered by inhibition of anti-activin A via monoclonal antibody treatment. Hence, we surmised that anti-ACVR1 antibodies that block activation of ACVR1 by ligands should also inhibit HO in FOP and provide an additional therapeutic option for this condition. Therefore, we generated anti-ACVR1 monoclonal antibodies that block ACVR1's activation by its ligands. Surprisingly, in vivo, these anti-ACVR1 antibodies stimulated HO and activated signaling of FOP-mutant ACVR1. This property was restricted to FOP-mutant ACVR1 and resulted from anti-ACVR1 antibody-mediated dimerization of ACVR1. Conversely, wild-type ACVR1 was inhibited by anti-ACVR1 antibodies. These results uncover an additional property of FOP-mutant ACVR1 and indicate that anti-ACVR1 antibodies should not be considered as therapeutics for FOP.


Subject(s)
Myositis Ossificans , Ossification, Heterotopic , Activin Receptors, Type I/genetics , Activin Receptors, Type I/pharmacology , Antibodies/immunology , Humans , Ligands , Mutation , Myositis Ossificans/genetics , Ossification, Heterotopic/genetics , Ossification, Heterotopic/pathology , Signal Transduction/genetics
2.
Elife ; 92020 06 09.
Article in English | MEDLINE | ID: mdl-32515349

ABSTRACT

Activin A functions in BMP signaling in two ways: it either engages ACVR1B to activate Smad2/3 signaling or binds ACVR1 to form a non-signaling complex (NSC). Although the former property has been studied extensively, the roles of the NSC remain unexplored. The genetic disorder fibrodysplasia ossificans progressiva (FOP) provides a unique window into ACVR1/Activin A signaling because in that disease Activin can either signal through FOP-mutant ACVR1 or form NSCs with wild-type ACVR1. To explore the role of the NSC, we generated 'agonist-only' Activin A muteins that activate ACVR1B but cannot form the NSC with ACVR1. Using one of these muteins, we demonstrate that failure to form the NSC in FOP results in more severe disease pathology. These results provide the first evidence for a biological role for the NSC in vivo and pave the way for further exploration of the NSC's physiological role in corresponding knock-in mice.


Subject(s)
Activin Receptors, Type I/metabolism , Activins/metabolism , Bone Morphogenetic Proteins/metabolism , Myositis Ossificans/genetics , Signal Transduction/genetics , Activin Receptors, Type I/genetics , Activins/genetics , Animals , Bone Morphogenetic Protein Receptors, Type II/genetics , Bone Morphogenetic Protein Receptors, Type II/metabolism , Bone Morphogenetic Proteins/genetics , Gene Knock-In Techniques , Mice , Mice, Transgenic , Mutation , Myositis Ossificans/pathology
3.
Bone ; 138: 115473, 2020 09.
Article in English | MEDLINE | ID: mdl-32553795

ABSTRACT

Heterotopic ossification (HO), the formation of ectopic bone in soft tissues, has been extensively studied in its two primary forms: post-traumatic HO (tHO) typically found in patients who have experienced musculoskeletal or neurogenic injury and in fibrodysplasia ossificans progressiva (FOP), where it is genetically driven. Given that in both diseases HO arises via endochondral ossification, the molecular mechanisms behind both diseases have been postulated to be manifestations of similar pathways including those activated by BMP/TGFß superfamily ligands. A significant step towards understanding the molecular mechanism by which HO arises in FOP was the discovery that FOP causing ACVR1 variants trigger HO in response to activin A, a ligand that does not activate signaling from wild type ACVR1, and that is not inherently osteogenic in wild type settings. The physiological significance of this finding was demonstrated by showing that activin A neutralizing antibodies stop HO in two different genetically accurate mouse models of FOP. In order to explore the role of activin A in tHO, we performed single cell RNA sequencing and compared the expression of activin A as well as other BMP pathway genes in tHO and FOP HO. We show that activin A is expressed in response to injury in both settings, but by different types of cells. Given that wild type ACVR1 does not transduce signal when engaged by activin A, we hypothesized that inhibition of activin A will not block tHO. Nonetheless, as activin A was expressed in tHO lesions, we tested its inhibition and compared it with inhibition of BMPs. We show here that anti-activin A does not block tHO, whereas agents such as antibodies that neutralize ACVR1 or ALK3-Fc (which blocks osteogenic BMPs) are beneficial, though not completely curative. These results demonstrate that inhibition of activin A should not be considered as a therapeutic strategy for ameliorating tHO.


Subject(s)
Myositis Ossificans , Ossification, Heterotopic , Activin Receptors, Type I/genetics , Activins , Animals , Humans , Mice , Myositis Ossificans/genetics
4.
Eur J Hum Genet ; 28(9): 1243-1264, 2020 09.
Article in English | MEDLINE | ID: mdl-32376988

ABSTRACT

Previously we reported the identification of a homozygous COL27A1 (c.2089G>C; p.Gly697Arg) missense variant and proposed it as a founder allele in Puerto Rico segregating with Steel syndrome (STLS, MIM #615155); a rare osteochondrodysplasia characterized by short stature, congenital bilateral hip dysplasia, carpal coalitions, and scoliosis. We now report segregation of this variant in five probands from the initial clinical report defining the syndrome and an additional family of Puerto Rican descent with multiple affected adult individuals. We modeled the orthologous variant in murine Col27a1 and found it recapitulates some of the major Steel syndrome associated skeletal features including reduced body length, scoliosis, and a more rounded skull shape. Characterization of the in vivo murine model shows abnormal collagen deposition in the extracellular matrix and disorganization of the proliferative zone of the growth plate. We report additional COL27A1 pathogenic variant alleles identified in unrelated consanguineous Turkish kindreds suggesting Clan Genomics and identity-by-descent homozygosity contributing to disease in this population. The hypothesis that carrier states for this autosomal recessive osteochondrodysplasia may contribute to common complex traits is further explored in a large clinical population cohort. Our findings augment our understanding of COL27A1 biology and its role in skeletal development; and expand the functional allelic architecture in this gene underlying both rare and common disease phenotypes.


Subject(s)
Abnormalities, Multiple/genetics , Fibrillar Collagens/genetics , Founder Effect , Hip Dislocation/genetics , Scoliosis/genetics , Abnormalities, Multiple/pathology , Adolescent , Animals , Bone Development , Child , Child, Preschool , Consanguinity , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Female , Fibrillar Collagens/metabolism , Gene Frequency , Hip Dislocation/pathology , Homozygote , Humans , Male , Mice , Mice, Inbred C57BL , Mutation , Pedigree , Scoliosis/pathology , Syndrome
5.
Int J Mol Sci ; 20(5)2019 Mar 12.
Article in English | MEDLINE | ID: mdl-30871004

ABSTRACT

Most breast cancer patients die due to bone metastasis. Although metastasis accounts for 5% of the breast cancer cases, it is responsible for most of the deaths. Sometimes even before the detection of a primary tumor, most of the patients have bone and lymph node metastasis. Moreover, at the time of death, breast cancer patients have the bulk of the tumor burden in their bones. Therapy options are available for the treatment of primary tumors, but there are minimal options for treating breast cancer patients who have bone metastasis. C-X-C motif chemokine receptor type 2 (CXCR2) receptor-mediated signaling has been shown to play a critical role during bone-related inflammations and its ligands C-X-C motif chemokine ligand 6 (CXCL6) and 8 (CXCL8) aid in the resorption of bone during bone metastasis. In this study, we tested the hypothesis that CXCR2 contributes to mammary tumor-induced osteolysis and bone metastasis. In the present study, we examined the role of both tumor cell-derived and host-derived CXCR2 in influencing mammary tumor cell bone metastasis. For understanding the role of tumor cell-derived CXCR2, we utilized Cl66 CXCR2 knockdown (Cl66-shCXCR2) and Cl66-Control cells (Cl66-Control) and observed a significant decrease in tumor growth and tumor-induced osteolysis in Cl66-shCXCR2 cells in comparison with the Cl66-Control cells. Next, for understanding the role of host-derived CXCR2, we utilized mice with genomic knockdown of CXCR2 (Cxcr2-/-) and injected Cl66-Luciferase (Cl66-Luc) or 4T1-Luciferase (4T1-Luc) cells. We observed decreased bone destruction and metastasis in the bone of Cxcr2-/- mice. Our data suggest the importance of both tumor cell- and host-derived CXCR2 signaling in the bone metastasis of breast cancer cells.


Subject(s)
Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Lymphatic Metastasis/pathology , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Receptors, Interleukin-8B/metabolism , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/physiology , Female , Inflammation/metabolism , Inflammation/pathology , Mice , Mice, Inbred BALB C , Osteolysis/metabolism , Osteolysis/pathology , Signal Transduction/physiology , Tumor Burden/physiology
6.
Sci Transl Med ; 7(303): 303ra137, 2015 Sep 02.
Article in English | MEDLINE | ID: mdl-26333933

ABSTRACT

Fibrodysplasia ossificans progressiva (FOP) is a rare genetic disorder characterized by episodically exuberant heterotopic ossification (HO), whereby skeletal muscle is abnormally converted into misplaced, but histologically normal bone. This HO leads to progressive immobility with catastrophic consequences, including death by asphyxiation. FOP results from mutations in the intracellular domain of the type I BMP (bone morphogenetic protein) receptor ACVR1; the most common mutation alters arginine 206 to histidine (ACVR1(R206H)) and has been thought to drive inappropriate bone formation as a result of receptor hyperactivity. We unexpectedly found that this mutation rendered ACVR1 responsive to the activin family of ligands, which generally antagonize BMP signaling through ACVR1 but cannot normally induce bone formation. To test the implications of this finding in vivo, we engineered mice to carry the Acvr1(R206H) mutation. Because mice that constitutively express Acvr1[R206H] die perinatally, we generated a genetically humanized conditional-on knock-in model for this mutation. When Acvr1[R206H] expression was induced, mice developed HO resembling that of FOP; HO could also be triggered by activin A administration in this mouse model of FOP but not in wild-type controls. Finally, HO was blocked by broad-acting BMP blockers, as well as by a fully human antibody specific to activin A. Our results suggest that ACVR1(R206H) causes FOP by gaining responsiveness to the normally antagonistic ligand activin A, demonstrating that this ligand is necessary and sufficient for driving HO in a genetically accurate model of FOP; hence, our human antibody to activin A represents a potential therapeutic approach for FOP.


Subject(s)
Activin Receptors, Type I/genetics , Activins/metabolism , Mutation , Myositis Ossificans/genetics , Activin Receptors, Type I/metabolism , Animals , Mice , Mice, Transgenic , Protein Binding , Tacrolimus Binding Protein 1A/metabolism
7.
Clin Exp Metastasis ; 32(1): 65-72, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25511644

ABSTRACT

Host-derived angiogenic and inflammatory tumor supportive microenvironment regulates progression and metastasis, but the molecular mechanism(s) underlying host-tumor interactions remains unclear. Tumor expression of CXCR2 and its ligands have been shown to regulate angiogenesis, invasion, tumor growth, and metastasis. In this report, we hypothesized that host-derived Cxcr2-dependent signaling plays an important role in breast cancer growth and metastasis. Two mammary tumor cell lines Cl66 and 4T1 cells were orthotopically implanted into the mammary fat pad of wild-type and Cxcr2(-/-) female BALB/c mice. Tumor growth and spontaneous lung metastasis were monitored. Immunohistochemical analyses of the tumor tissues were performed to analyze proliferation, angiogenesis, apoptosis and immune cell infiltration. Our results demonstrated that knock-down of host Cxcr2 decreases tumor growth and metastasis by reducing angiogenesis, proliferation and enhancing apoptosis. Host Cxcr2 plays an important role in governing the pro-inflammatory response in mammary tumors as evaluated by decreased Gr1(+) tumor-associated granulocytes, F4/80(+) tumor associated macrophages, and CD11b(+)Gr1(+) myeloid derived suppressor cells in Cxcr2(-/-) mice as compared to control wild-type mice. Together, these results demonstrate that host Cxcr2-dependent signaling regulates mammary tumor growth and metastasis by promoting angiogenesis and pro-inflammatory responses.


Subject(s)
Apoptosis/genetics , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/genetics , Neovascularization, Pathologic/genetics , Receptors, Interleukin-8B/biosynthesis , Animals , Antigens, Differentiation/metabolism , CD11b Antigen/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Female , Granulocytes/immunology , Inflammation/immunology , Macrophages/immunology , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Neutrophils/immunology , Receptors, Interleukin-8B/genetics , Signal Transduction , Tumor Microenvironment/immunology
8.
Mol Cancer Ther ; 12(5): 799-808, 2013 May.
Article in English | MEDLINE | ID: mdl-23468530

ABSTRACT

Breast cancer is one of the leading causes of cancer deaths among females. Many challenges exist in the current management of advanced stage breast cancer as there are fewer recognized therapeutic strategies, often because of therapy resistance. How breast cancer cells evade chemotherapy and the underlying mechanism remains unclear. We and others have observed that malignant cells that survive initial chemo- and radiation therapy express higher levels of CXCR2 ligands, which may provide a survival benefit leading to therapy resistance. In this report, we test the hypothesis that CXCR2-dependent signaling in malignant cells may be critical for chemotherapy resistance and targeting this signaling axis may enhance the antitumor and antimetastatic activity of chemotherapeutic drugs and limit their toxicity. We used Cl66-wt, 4T1-wt, Cl66sh-CXCR2, and 4T1sh-CXCR2 cells expressing differential levels of the CXCR2 receptor to evaluate the role of targeting CXCR2 on chemotherapeutic responses. Knockdown of CXCR2 enhances paclitaxel and doxorubicin-mediated toxicity at suboptimal doses. Moreover, we observed an increase in the expression of CXCL1, a CXCR2 ligand in paclitaxel and doxorubicin-treated mammary tumor cells, which were inhibited following CXCR2 knockdown. Knockdown of CXCR2 enhanced antitumor activity of paclitaxel in an in vivo mammary tumor model. We observed significant inhibition of spontaneous lung metastases in animals bearing CXCR2 knockdown tumors and treated with paclitaxel as compared with the control group. Our data suggest the novel role of CXCR2 and its ligands in maintaining chemotherapy resistance and provide evidence that targeting CXCR2 signaling in an adjuvant setting will help circumvent chemotherapy resistance.


Subject(s)
Antineoplastic Agents/pharmacology , Lung Neoplasms/genetics , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Neovascularization, Pathologic/genetics , Receptors, Interleukin-8B/genetics , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Ligands , Mammary Neoplasms, Experimental/drug therapy , Mice , Neovascularization, Pathologic/drug therapy , Paclitaxel/administration & dosage , Paclitaxel/pharmacology , Tumor Burden/drug effects , Tumor Burden/genetics
9.
J Carcinog ; 10: 40, 2011.
Article in English | MEDLINE | ID: mdl-22368515

ABSTRACT

BACKGROUND: Chemokines and their receptors have long been known to regulate metastasis in various cancers. Previous studies have shown that CXCR2 expression is upregulated in malignant breast cancer tissues but not in benign ductal epithelial samples. The functional role of CXCR2 in the metastatic phenotype of breast cancer still remains unclear. We hypothesize that the chemokine receptor, CXCR2, mediates tumor cell invasion and migration and promotes metastasis in breast cancer. The objective of this study is to investigate the potential role of CXCR2 in the metastatic phenotype of mouse mammary tumor cells. MATERIALS AND METHODS: We evaluated the functional role of CXCR2 in breast cancer by stably downregulating the expression of CXCR2 in metastatic mammary tumor cell lines Cl66 and 4T1, using short hairpin RNA (shRNA). The effects of CXCR2 downregulation on tumor growth, invasion and metastatic potential were analyzed in vitro and in vivo. RESULTS: We demonstrated knock down of CXCR2 in Cl66 and 4T1 cells (Cl66-shCXCR2 and 4T1-shCXCR2) cells by reverse transcriptase polymerase chain reaction (RT-PCR) at the transcriptional level and by immunohistochemistry at the protein level. We did not observe a significant difference in in vitro cell proliferation between vector control and CXCR2 knock-down Cl66 or 4T1 cells. Next, we examined the invasive potential of Cl66-shCXCR2 cells by in vitro Matrigel invasion assay. We observed a significantly lower number (52 ± 5) of Cl66-shCXCR2 cells invading through Matrigel compared to control cells (Cl66-control) (182 ± 3) (P < 0.05). We analyzed the in vivo metastatic potential of Cl66-shCXCR2 using a spontaneous metastasis model by orthotopically implanting cells into the mammary fat pad of female BALB/c mice. Animals were sacrificed 12 weeks post tumor implantation and tissue samples were analyzed for metastatic nodules. CXCR2 downregulation significantly inhibited tumor cell metastasis. All the mice (n = 10) implanted with control Cl66 cells spontaneously developed lung metastasis, whereas a significantly lower number of mice (40%) implanted with Cl66-shCXCR2 cells exhibited lung metastases. CONCLUSIONS: Together, these results suggest that CXCR2 may play a critical role in breast cancer invasion and metastasis.

10.
Curr Osteoporos Rep ; 8(2): 105-13, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20425618

ABSTRACT

The metastasis of tumor cells to distant organs is the primary cause of cancer-related mortality in most cancers. The interaction of tumor cells with local stroma at the metastatic site plays a critical role in metastatic dissemination and the establishment of metastases. These tumor-stromal interactions regulate several important steps including degradation of extracellular matrix, release of sequestered growth factors, and expression of chemokines, cytokines, and receptors on tumor cells and the interacting stromal cells. Breast, prostate, and lung cancers preferentially metastasize to bone. Tumor cell interactions with the bone microenvironment initiate a series of complex cellular interactions that promotes establishment of osteoclastic and/or osteoblastic metastasis. Understanding the interactions between tumor cells and the stroma is important to identify molecular targets to develop novel therapies aimed at reducing metastasis formation. In this article, we review the important mechanisms of tumor-stromal interaction in the development of bone metastasis.


Subject(s)
Bone Neoplasms/secondary , Cell Communication , Osteoblasts/pathology , Bone Neoplasms/pathology , Disease Progression , Humans , Neoplasm Metastasis
11.
Cancer Res ; 70(9): 3494-504, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20406980

ABSTRACT

The tropism of breast cancer cells for bone and their tendency to induce an osteolytic phenotype are a result of interactions between breast cancer cells and stromal cells and are of paramount importance for bone metastasis. However, the underlying molecular mechanisms remain poorly understood. We hypothesize that tumor-stromal interaction alters gene expression in malignant tumor cells and stromal cells creating a unique expression signature that promotes osteolytic breast cancer bone metastasis and that inhibition of such interactions can be developed as targeted therapeutics. Microarray analysis was performed to investigate gene expression profiling at the tumor-bone (TB) interface versus the tumor alone area from syngenic mice injected with three different syngenic mammary tumor cell lines that differ in their metastatic potential. We identified matrix metalloproteinase 13 (MMP13), receptor activator of NF-kappaB ligand (RANKL), and integrins binding sialoprotein to be genes upregulated at the TB interface and validated. To determine the functional role of MMP13 in tumor-induced osteolysis, mice with Cl66 mammary tumors were treated with MMP13 antisense oligonucleotides (MMP13-ASO) or control scrambled oligonucleotides (control-ASO). Knockdown of MMP13 expression at the TB interface leads to significant reduction in bone destruction and in the number of activated osteoclasts at the TB interface. Further analysis to evaluate the mechanism of MMP13-dependent osteolytic bone metastasis revealed that MMP13-ASO treatment decreased active MMP9, RANKL levels, and transforming growth factor-beta signaling at the TB interface. Together, our data indicate that upregulation of MMP13 at the TB interface is important in tumor-induced osteolysis and suggest that MMP13 is a potential therapeutic target for breast cancer bone metastasis.


Subject(s)
Bone Neoplasms/secondary , Mammary Neoplasms, Experimental/enzymology , Mammary Neoplasms, Experimental/pathology , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 9/metabolism , Osteolysis/enzymology , Transforming Growth Factor beta/metabolism , Adenocarcinoma/enzymology , Adenocarcinoma/pathology , Animals , Bone Neoplasms/enzymology , Bone Neoplasms/metabolism , Bone and Bones/enzymology , Bone and Bones/metabolism , Bone and Bones/pathology , Cell Line, Tumor , Enzyme Activation , Female , Gene Expression Profiling , Mammary Neoplasms, Experimental/metabolism , Matrix Metalloproteinase 13/biosynthesis , Matrix Metalloproteinase 13/genetics , Matrix Metalloproteinase 9/biosynthesis , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase Inhibitors , Mice , Mice, Inbred BALB C , Osteoclasts/enzymology , Osteoclasts/pathology , Osteolysis/genetics , Osteolysis/metabolism , Osteolysis/pathology , Osteoprotegerin/biosynthesis , RANK Ligand/biosynthesis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Signal Transduction , Up-Regulation
12.
Int J Cancer ; 126(2): 328-36, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-19585580

ABSTRACT

CXCR1 and CXCR2 are receptors for CXCL-8 and are differentially expressed on melanoma and endothelial cells. In this study, we determined the functional role of these receptors in melanoma progression. We stably knock-down the expression of CXCR1 and/or CXCR2 in A375-SM (SM; high metastatic) human melanoma cells by short-hairpin RNA transfection. Cell proliferation, migration, invasion, ERK phosphorlyation and cytoskeletal rearrangements were carried out in vitro. In vivo growth was evaluated using murine subcutaneous xenograft model. Our data demonstrate that knock-down of CXCR1 and/or CXCR2 expression, inhibited melanoma cell proliferation, survival, migration and invasive potential in vitro. Moreover, we also observed inhibition of ERK phosphorylation and cytoskeltal rearrangement in SM-shCXCR1, SM-shCXCR2 and SM-shCXCR1/2 cells. Furthermore, when SM-shCXCR1 or SM-shCXCR2 cells implanted in nude mice, tumor growth, proliferation and microvessel density was significantly inhibited as compared to SM-control cells. In addition, we observed a significant increase in melanoma cell apoptosis in SM-shCXCR1 and SM-shCXCR2 tumors compared to SM-control tumors. Together, these data demonstrate that CXCR1 and CXCR2 expression play a critical role in human melanoma tumor progression and, functional blockade of CXCR1 and CXCR2 could be potentially used for future therapeutic intervention in malignant melanoma.


Subject(s)
Melanoma/pathology , RNA Interference , Receptors, Interleukin-8A/physiology , Receptors, Interleukin-8B/physiology , Actins/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , Blotting, Western , Cell Line, Tumor , Cell Proliferation , Cell Survival/genetics , Cell Survival/physiology , Female , Humans , Melanoma/blood supply , Melanoma/genetics , Melanoma, Experimental/blood supply , Melanoma, Experimental/genetics , Melanoma, Experimental/pathology , Mice , Mice, Nude , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neoplasm Invasiveness , Neoplasm Transplantation , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Phosphorylation , RNA, Small Interfering/genetics , Receptors, Interleukin-8A/genetics , Receptors, Interleukin-8A/metabolism , Receptors, Interleukin-8B/genetics , Receptors, Interleukin-8B/metabolism , Transplantation, Heterologous , Tumor Burden
13.
Cancer Lett ; 288(2): 162-9, 2010 Feb 28.
Article in English | MEDLINE | ID: mdl-19646811

ABSTRACT

Transforming growth factor (TGF)-beta signaling makes a significant contribution to the pathogenesis of breast cancer bone metastasis. In other tumor types, TGF-beta has been shown to promote tumor vascularity. Here, we report that inhibition of TGF-beta significantly reduces microvessel density in mammary tumor-induced bone lesions, mediated by decreased expression of both vascular endothelial growth factor (VEGF) and monocyte chemotactic protein (MCP)-1, both known angiogenic factors. Cathepsin G upregulation at the tumor-bone interface has been linked to increased TGF-beta signaling, and we also report that inhibition of Cathepsin G reduced tumor vascularity, as well as VEGF and MCP-1 expression.


Subject(s)
Bone Neoplasms/enzymology , Breast Neoplasms/pathology , Cathepsin G/metabolism , Chemokine CCL2/metabolism , Neovascularization, Pathologic/enzymology , Signal Transduction , Transforming Growth Factor beta/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Antibodies , Bone Neoplasms/blood supply , Bone Neoplasms/drug therapy , Bone Neoplasms/secondary , Cathepsin G/antagonists & inhibitors , Cell Line, Tumor , Chemokine CCL2/genetics , Female , Gene Expression Regulation, Neoplastic , Mice , Mice, Inbred BALB C , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/prevention & control , RNA, Messenger/metabolism , Serine Proteinase Inhibitors/pharmacology , Signal Transduction/drug effects , Tosylphenylalanyl Chloromethyl Ketone/pharmacology , Transforming Growth Factor beta/immunology , Up-Regulation , Vascular Endothelial Growth Factor A/genetics
14.
Mol Cancer Res ; 7(8): 1224-33, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19671689

ABSTRACT

Increased transforming growth factor-beta (TGF-beta) signaling has been observed at the tumor-bone interface of mammary tumor-induced osteolytic lesions despite no observed transcriptional up-regulation of TGF-beta. To this point, the mechanism for enhanced TGF-beta signaling remains unclear. The bulk of TGF-beta that is released at the tumor-bone interface is in an inactive form secondary to association with beta-latency-associated protein and latency TGF-beta binding protein. We hypothesized that the observed increase in TGF-beta signaling is due to increased cathepsin G-dependent, matrix metalloproteinase 9 (MMP9)-mediated activation of latent TGF-beta. MMP9 is capable of activating latent TGF-beta, and we observed that decreased production of MMP9 was associated with reduced TGF-beta signaling. Similar to TGF-beta, MMP9 is released in an inactive form and requires proteolytic activation. We showed that cathepsin G, which we have previously shown to be up-regulated at the tumor-bone interface, is capable of activating pro-MMP9. Inhibition of cathepsin G in vivo significantly reduced MMP9 activity, increased the ratio of latent TGF-beta to active TGF-beta, and reduced the level of TGF-beta signaling. Our proposed model based on these results is that cathepsin G is up-regulated through tumor-stromal interactions and activates pro-MMP9, active MMP9 cleaves and releases active TGF-beta, and active TGF-beta can then promote tumor growth and enhance osteoclast activation and subsequent bone resorption. Thus, for the first time, we have identified cathepsin G and MMP9 as proteases involved in enhanced TGF-beta signaling at the tumor-bone interface of mammary tumor-induced osteolytic lesions and have identified these proteases as potential therapeutic targets.


Subject(s)
Bone Neoplasms/enzymology , Bone Neoplasms/pathology , Bone and Bones/pathology , Cathepsin G/metabolism , Enzyme Precursors/metabolism , Matrix Metalloproteinase 9/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Animals , Bone Neoplasms/complications , Bone and Bones/drug effects , Bone and Bones/enzymology , Cathepsin G/antagonists & inhibitors , Enzyme Activation/drug effects , Mammary Tumor Virus, Mouse , Matrix Metalloproteinase 2/metabolism , Mice , Oligonucleotides, Antisense/pharmacology , Osteolysis/complications , Osteolysis/enzymology , Osteolysis/pathology , RANK Ligand/metabolism , Signal Transduction/drug effects
15.
Clin Exp Metastasis ; 26(7): 797-808, 2009.
Article in English | MEDLINE | ID: mdl-19590968

ABSTRACT

The bone microenvironment plays a critical role in tumor-induced osteolysis and osteolytic metastasis through tumor-bone (TB)-interaction. Receptor activator of nuclear factor-kappaB (RANK) ligand (RANKL) is one of the critical signaling molecules involved in osteolysis and bone metastasis. However, the regulation and functional significance of RANKL at the TB-interface in tumor-induced osteolysis remains unclear. In this report, we examined the role of tumor-stromal interaction in the regulation of RANKL expression and its functional significance in tumor-induced osteolysis. Using a novel mammary tumor model, we identified that RANKL expression was upregulated at the TB-interface as compared to the tumor alone area. We demonstrate increased generation of sRANKL at the TB-interface, which is associated with tumor-induced osteolysis. The ratio of RANKL to osteoprotegrin (OPG), a decoy receptor for RANKL, at the TB-interface was also increased. Targeting RANKL expression with antisense oligonucleotides (RANKL-ASO), significantly abrogated tumor-induced osteolysis, decreased RANKL expression and the RANKL:OPG ratio at the TB-interface. Together, these results demonstrate that upregulation of RANKL expression and sRANKL generation at the TB-interface potentiates tumor-induced osteolysis.


Subject(s)
Bone and Bones/metabolism , Mammary Neoplasms, Experimental/metabolism , RANK Ligand/metabolism , Animals , Base Sequence , Blotting, Western , Bone and Bones/pathology , DNA Primers , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Ligands , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Osteolysis , Reverse Transcriptase Polymerase Chain Reaction
16.
Cancer Res ; 69(7): 3188-95, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19293192

ABSTRACT

Metastatic breast cancer shows extreme tropism for the bone microenvironment, leading to the establishment of osteolytic metastases. Perpetuation of tumor-induced osteolysis requires a continuous supply of osteoclast precursors migrating into the bone microenvironment that can subsequently differentiate into mature osteoclasts and resorb bone. Thus, identification and subsequent targeting of chemoattractants of osteoclast precursors that are up-regulated at the tumor-bone interface represents a potential avenue to interrupt osteolysis. We report that cathepsin G, a serine protease, plays a vital role in the bone microenvironment by modulating tumor-stromal interaction in a manner that favors tumor establishment and regulates chemotaxis of monocytes, a subset of which has the potential to differentiate into osteoclasts. Our data show that cathepsin G-induced chemotaxis of monocytes is mediated by proteolytic activation of protease-activated receptor-1 (PAR-1). Attenuation of PAR-1 activation abrogates cathepsin G-mediated induction of monocyte chemotaxis. We also show that in vivo inhibition of cathepsin G reduces the number of CD11b(+) osteoclast precursors and mature osteoclasts at the tumor-bone interface. Together, these data suggest that therapeutic targeting of both PAR-1 signaling in osteoclast precursors as well as cathepsin G at the tumor-bone interface has the potential to reduce osteolysis by inhibiting the recruitment, differentiation, and activation of osteoclast precursors.


Subject(s)
Cathepsins/metabolism , Osteoclasts/metabolism , Receptor, PAR-1/metabolism , Serine Endopeptidases/metabolism , Animals , Cathepsin G , Cathepsins/antagonists & inhibitors , Cathepsins/biosynthesis , Cathepsins/pharmacology , Cell Line , Chemotaxis/drug effects , Female , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Humans , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Mammary Neoplasms, Experimental/enzymology , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Osteoclasts/cytology , Osteoclasts/drug effects , Osteoclasts/pathology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptor, PAR-1/biosynthesis , Receptors, Thrombin/biosynthesis , Serine Endopeptidases/biosynthesis , Serine Endopeptidases/pharmacology , Signal Transduction , Tosylphenylalanyl Chloromethyl Ketone/pharmacology
17.
Clin Cancer Res ; 15(7): 2380-6, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19293256

ABSTRACT

PURPOSE: Melanoma, the most aggressive form of skin cancer, accounts for 75% of all skin cancer-related deaths and current therapeutic strategies are not effective in advanced disease. In the current study, we have investigated the efficacy of orally active small-molecule antagonist targeting CXCR2/CXCR1. EXPERIMENTAL DESIGN: Human A375SM melanoma cells were treated with SCH-479833 or SCH-527123, and their effect on proliferation, motility, and invasion was evaluated in vitro. We examined the downstream signaling events in the cells following treatment with antagonists. For in vivo studies, A375SM cells were implanted subcutaneously into athymic nude mice followed by administration of SCH-479833, SCH-527123, or hydroxypropyl-beta-cyclodextrin (20%) orally for 21 days and their effect on tumor growth and angiogenesis was evaluated. RESULTS: Our data show that SCH-479833 or SCH-527123 inhibited the melanoma cell proliferation, chemotaxis, and invasive potential in vitro. Treatment of melanoma cells with SCH-479833 or SCH-527123 also inhibited tumor growth. Histologic and histochemical analyses showed significant (P < 0.05) decreases in tumor cell proliferation and microvessel density in tumors. Moreover, we observed a significant increase in melanoma cell apoptosis in SCH-479833- or SCH-527123-treated animals compared with controls. CONCLUSION: Together, these studies show that selectively targeting CXCR2/CXCR1 with orally active small-molecule inhibitors is a promising therapeutic approach for inhibiting melanoma growth and angiogenesis.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Benzamides/therapeutic use , Cyclobutanes/therapeutic use , Melanoma/drug therapy , Receptors, Interleukin-8A/antagonists & inhibitors , Receptors, Interleukin-8B/antagonists & inhibitors , Skin Neoplasms/drug therapy , Administration, Oral , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/chemistry , Animals , Benzamides/administration & dosage , Benzamides/chemistry , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation , Cell Survival , Chemotaxis/drug effects , Cyclobutanes/administration & dosage , Cyclobutanes/chemistry , Female , Humans , MAP Kinase Signaling System/drug effects , Melanoma/blood supply , Melanoma/pathology , Mice , Mice, Nude , Neovascularization, Pathologic/drug therapy , Skin Neoplasms/blood supply , Skin Neoplasms/pathology
18.
Cancer Sci ; 100(1): 71-81, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19038005

ABSTRACT

Understanding the cellular and molecular changes in the bone microenvironment is important for developing novel therapeutics to control breast cancer bone metastasis. Although the underlying mechanism(s) of bone metastasis has been the focus of intense investigation, relatively little is known about complex molecular interactions between malignant cells and bone stroma. Using a murine syngeneic model that mimics osteolytic changes associated with human breast cancer, we examined the role of tumor-bone interaction in tumor-induced osteolysis and malignant growth in the bone microenvironment. We identified transforming growth factor-beta receptor 1 (TGF-betaRI) as a commonly upregulated gene at the tumor-bone (TB) interface. Moreover, TGF-betaRI expression and activation, analyzed by nuclear localization of phospho-Smad2, was higher in tumor cells and osteoclasts at the TB interface as compared to the tumor-alone area. Furthermore, attenuation of TGF-beta activity by neutralizing antibody to TGF-beta or TGF-betaRI kinase inhibitor reduced mammary tumor-induced osteolysis, TGF-betaRI expression and its activation. In addition, we demonstrate a potential role of TGF-beta as an important modifier of receptor activator of NF-kappaB ligand (RANKL)-dependent osteoclast activation and osteolysis. Together, these studies demonstrate that inhibition of TGF-betaRI signaling at the TB interface will be a therapeutic target in the treatment of breast cancer-induced osteolysis.


Subject(s)
Bone Neoplasms/secondary , Mammary Neoplasms, Experimental/pathology , Osteoclasts/physiology , Signal Transduction/physiology , Transforming Growth Factor beta/physiology , Animals , Cell Differentiation , Cell Line, Tumor , Female , Mice , Mice, Inbred BALB C , Osteoclasts/cytology , Osteolysis , Protein Serine-Threonine Kinases/analysis , Protein Serine-Threonine Kinases/genetics , RANK Ligand/physiology , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/analysis , Receptors, Transforming Growth Factor beta/genetics
19.
Cancer Res ; 68(14): 5803-11, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18632634

ABSTRACT

Breast cancer commonly causes osteolytic metastases in bone, a process that is dependent on tumor-stromal interaction. Proteases play an important role in modulating tumor-stromal interactions in a manner that favors tumor establishment and progression. Whereas several studies have examined the role of proteases in modulating the bone microenvironment, little is currently known about their role in tumor-bone interaction during osteolytic metastasis. In cancer-induced osteolytic lesions, cleavage of receptor activator of nuclear factor-kappaB ligand (RANKL) to a soluble version (sRANKL) is critical for widespread osteoclast activation. Using a mouse model that mimics osteolytic changes associated with breast cancer-induced bone metastases, we identified cathepsin G, cathepsin K, matrix metalloproteinase (MMP)-9, and MMP13 to be proteases that are up-regulated at the tumor-bone interface using comparative cDNA microarray analysis and quantitative reverse transcription-PCR. Moreover, we showed that cathepsin G is capable of shedding the extracellular domain of RANKL, generating active sRANKL that is capable of inducing differentiation and activation of osteoclast precursors. The major source of cathepsin G at the tumor-bone interface seems to be osteoclasts that up-regulate production of cathepsin G via interaction with tumor cells. Furthermore, we showed that in vitro osteoclastogenesis is reduced by inhibition of cathepsin G in a coculture model and that in vivo inhibition of cathepsin G reduces mammary tumor-induced osteolysis. Together, our data indicate that cathepsin G activity at the tumor-bone interface plays an important role in mammary tumor-induced osteolysis and suggest that cathepsin G is a potentially novel therapeutic target in the treatment of breast cancer bone metastasis.


Subject(s)
Bone Neoplasms/secondary , Cathepsins/physiology , Mammary Neoplasms, Animal/metabolism , NF-kappa B/metabolism , Osteolysis , RANK Ligand/metabolism , Serine Endopeptidases/physiology , Animals , Bone Neoplasms/pathology , Cathepsin G , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred BALB C , Neoplasm Metastasis , Neoplasm Transplantation , Osteoclasts/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...