Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Cell ; 150(1): 165-78, 2012 Jul 06.
Article in English | MEDLINE | ID: mdl-22770218

ABSTRACT

Metastasis and chemoresistance in cancer are linked phenomena, but the molecular basis for this link is unknown. We uncovered a network of paracrine signals between carcinoma, myeloid, and endothelial cells that drives both processes in breast cancer. Cancer cells that overexpress CXCL1 and 2 by transcriptional hyperactivation or 4q21 amplification are primed for survival in metastatic sites. CXCL1/2 attract CD11b(+)Gr1(+) myeloid cells into the tumor, which produce chemokines including S100A8/9 that enhance cancer cell survival. Although chemotherapeutic agents kill cancer cells, these treatments trigger a parallel stromal reaction leading to TNF-α production by endothelial and other stromal cells. TNF-α via NF-kB heightens the CXCL1/2 expression in cancer cells, thus amplifying the CXCL1/2-S100A8/9 loop and causing chemoresistance. CXCR2 blockers break this cycle, augmenting the efficacy of chemotherapy against breast tumors and particularly against metastasis. This network of endothelial-carcinoma-myeloid signaling interactions provides a mechanism linking chemoresistance and metastasis, with opportunities for intervention.


Subject(s)
Breast Neoplasms/pathology , Carcinoma/pathology , Chemokine CXCL1/metabolism , Drug Resistance, Neoplasm , Neoplasm Metastasis , Paracrine Communication , Animals , Breast Neoplasms/metabolism , Calgranulin A/metabolism , Calgranulin B/metabolism , Carcinoma/metabolism , Chemokine CXCL1/genetics , Disease Models, Animal , Endothelial Cells/metabolism , Female , Gene Knockdown Techniques , Humans , Lung Neoplasms/secondary , Lymph Nodes/pathology , Lymphatic Metastasis , Mice , Mice, Inbred C57BL , Myeloid Cells/metabolism , Neoplasm Transplantation , Transplantation, Heterologous
2.
Cell ; 139(7): 1315-26, 2009 Dec 24.
Article in English | MEDLINE | ID: mdl-20064377

ABSTRACT

Cancer cells that leave the primary tumor can seed metastases in distant organs, and it is thought that this is a unidirectional process. Here we show that circulating tumor cells (CTCs) can also colonize their tumors of origin, in a process that we call "tumor self-seeding." Self-seeding of breast cancer, colon cancer, and melanoma tumors in mice is preferentially mediated by aggressive CTCs, including those with bone, lung, or brain-metastatic tropism. We find that the tumor-derived cytokines IL-6 and IL-8 act as CTC attractants whereas MMP1/collagenase-1 and the actin cytoskeleton component fascin-1 are mediators of CTC infiltration into mammary tumors. We show that self-seeding can accelerate tumor growth, angiogenesis, and stromal recruitment through seed-derived factors including the chemokine CXCL1. Tumor self-seeding could explain the relationships between anaplasia, tumor size, vascularity and prognosis, and local recurrence seeded by disseminated cells following ostensibly complete tumor excision.


Subject(s)
Melanoma/pathology , Neoplasms/pathology , Neoplastic Cells, Circulating/pathology , Animals , Breast Neoplasms/pathology , Humans , Mice , Mice, Inbred BALB C , Neoplasm Proteins/metabolism , Neoplasm Recurrence, Local , Neoplasms/physiopathology , Prognosis , Skin Neoplasms/genetics
3.
EMBO Mol Med ; 16(8): 1957-1980, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38956205

ABSTRACT

Acquired resistance to PARP inhibitors (PARPi) remains a treatment challenge for BRCA1/2-mutant breast cancer that drastically shortens patient survival. Although several resistance mechanisms have been identified, none have been successfully targeted in the clinic. Using new PARPi-resistance models of Brca1- and Bard1-mutant breast cancer generated in-vivo, we identified FLT1 (VEGFR1) as a driver of resistance. Unlike the known role of VEGF signaling in angiogenesis, we demonstrate a novel, non-canonical role for FLT1 signaling that protects cancer cells from PARPi in-vivo through a combination of cell-intrinsic and cell-extrinsic pathways. We demonstrate that FLT1 blockade suppresses AKT activation, increases tumor infiltration of CD8+ T cells, and causes dramatic regression of PARPi-resistant breast tumors in a T-cell-dependent manner. Moreover, PARPi-resistant tumor cells can be readily re-sensitized to PARPi by targeting Flt1 either genetically (Flt1-suppression) or pharmacologically (axitinib). Importantly, a retrospective series of breast cancer patients treated with PARPi demonstrated shorter progression-free survival in cases with FLT1 activation at pre-treatment. Our study therefore identifies FLT1 as a potential therapeutic target in PARPi-resistant, BRCA1/2-mutant breast cancer.


Subject(s)
Breast Neoplasms , Drug Resistance, Neoplasm , Poly(ADP-ribose) Polymerase Inhibitors , Vascular Endothelial Growth Factor Receptor-1 , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Drug Resistance, Neoplasm/genetics , Humans , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-1/genetics , Female , Animals , Cell Line, Tumor , Mice , BRCA1 Protein/metabolism , BRCA1 Protein/genetics , Signal Transduction/drug effects
4.
Breast Cancer Res ; 15(2): 309, 2013 Apr 23.
Article in English | MEDLINE | ID: mdl-23634980

ABSTRACT

HER2 amplification and overexpression is observed in approximately 20% of breast cancers and is strongly associated with poor prognosis and therapeutic responsiveness to HER2 targeted agents. A recent study by Bose and colleagues suggests that another subset of breast cancer patients without HER2 amplification but with activating HER2 mutation might also benefit from existing HER2-targeted agents and the authors functionally characterize these somatic mutations in experimental models. In a second study on HER2-driven breast cancer, Angelini and colleagues investigate how the constitutively active, truncated carboxy-terminal fragment of HER2, p95HER2, promotes metastatic progression through non-cellautonomous secretion of factors from senescent cells. These new findings advance our understanding of HER2 biology in the context of HER2 activation as well as offer new insights into our understanding of drug sensitivity and metastatic progression.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/secondary , Cell Communication , Cellular Senescence/physiology , Drug Resistance, Neoplasm/genetics , Mutation/genetics , Receptor, ErbB-2/metabolism , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Disease Progression , Female , Humans , Receptor, ErbB-2/genetics
5.
Cancer Cell ; 8(5): 421-32, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16286249

ABSTRACT

Cachexia contributes to nearly a third of all cancer deaths, yet the mechanisms underlying skeletal muscle wasting in this syndrome remain poorly defined. We report that tumor-induced alterations in the muscular dystrophy-associated dystrophin glycoprotein complex (DGC) represent a key early event in cachexia. Muscles from tumor-bearing mice exhibited membrane abnormalities accompanied by reduced levels of dystrophin and increased glycosylation on DGC proteins. Wasting was accentuated in tumor mdx mice lacking a DGC but spared in dystrophin transgenic mice that blocked induction of muscle E3 ubiquitin ligases. Furthermore, DGC deregulation correlated positively with cachexia in patients with gastrointestinal cancers. Based on these results, we propose that, similar to muscular dystrophy, DGC dysfunction plays a critical role in cancer-induced wasting.


Subject(s)
Cachexia/metabolism , Dystrophin-Associated Proteins/physiology , Dystrophin/physiology , Glycoproteins/physiology , Neoplasms/metabolism , Adult , Aged , Animals , Cachexia/complications , Dystrophin/metabolism , Dystrophin-Associated Proteins/metabolism , Female , Founder Effect , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred mdx , Mice, Transgenic , Middle Aged , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Neoplasms/complications , Neoplasms/pathology , Ubiquitin-Protein Ligases/metabolism
6.
Nat Commun ; 13(1): 6095, 2022 10 15.
Article in English | MEDLINE | ID: mdl-36241629

ABSTRACT

We previously identified a chemotherapy-induced paracrine inflammatory loop that paradoxically mitigates the anti-tumor effect of chemotherapy and triggers metastatic propagation in breast and lung cancer models. Therefore, we sought to further validate and translate these findings into patient care by coupling the anti-TNF-α drug certolizumab pegol with standard cisplatin doublet chemotherapy. Here we first validate the anti-metastatic effect of certolizumab in a liver-metastatic Lewis Lung Carcinoma model. We then evaluate the safety, efficacy, and pharmacodynamic effects of certolizumab with cisplatin and pemetrexed in an open label Phase 1 clinical trial (NCT02120807) of eighteen adult patients with stage IV lung adenocarcinomas. The primary outcome is maximum tolerated dose. Secondary outcomes are response rate and progression-free survival (PFS); pharmacodynamic changes in blood and tumor are evaluated as a correlative outcome. There were nine partial responses among 16 patients evaluable (56%, 95% CI 30 to 80%). The median duration of response was 9.0 months (range 5.9 to 42.6 months) and median PFS was 7.1 months (95% CI 6.3 to NR). The standard 400 mg dose of certolizumab, added to cisplatin and pemetrexed, is well-tolerated and, as a correlative endpoint, demonstrates potent pharmacodynamic inhibition of peripheral cytokines associated with the paracrine inflammatory loop.


Subject(s)
Adenocarcinoma of Lung , Antineoplastic Combined Chemotherapy Protocols , Lung Neoplasms , Adult , Humans , Adenocarcinoma of Lung/drug therapy , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Certolizumab Pegol/therapeutic use , Cisplatin/therapeutic use , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Pemetrexed/therapeutic use , Tumor Necrosis Factor Inhibitors , Tumor Necrosis Factor-alpha/therapeutic use
7.
Cancer Discov ; 12(4): 1002-1021, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35078784

ABSTRACT

The epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) osimertinib has significantly prolonged progression-free survival (PFS) in patients with EGFR-mutant lung cancer, including those with brain metastases. However, despite striking initial responses, osimertinib-treated patients eventually develop lethal metastatic relapse, often to the brain. Although osimertinib-refractory brain relapse is a major clinical challenge, its underlying mechanisms remain poorly understood. Using metastatic models of EGFR-mutant lung cancer, we show that cancer cells expressing high intracellular S100A9 escape osimertinib and initiate brain relapses. Mechanistically, S100A9 upregulates ALDH1A1 expression and activates the retinoic acid (RA) signaling pathway in osimertinib-refractory cancer cells. We demonstrate that the genetic repression of S100A9, ALDH1A1, or RA receptors (RAR) in cancer cells, or treatment with a pan-RAR antagonist, dramatically reduces brain metastasis. Importantly, S100A9 expression in cancer cells correlates with poor PFS in osimertinib-treated patients. Our study, therefore, identifies a novel, therapeutically targetable S100A9-ALDH1A1-RA axis that drives brain relapse. SIGNIFICANCE: Treatment with the EGFR TKI osimertinib prolongs the survival of patients with EGFR-mutant lung cancer; however, patients develop metastatic relapses, often to the brain. We identified a novel intracellular S100A9-ALDH1A1-RA signaling pathway that drives lethal brain relapse and can be targeted by pan-RAR antagonists to prevent cancer progression and prolong patient survival. This article is highlighted in the In This Issue feature, p. 873.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Aldehyde Dehydrogenase 1 Family , Aniline Compounds/pharmacology , Aniline Compounds/therapeutic use , Brain/pathology , Carcinoma, Non-Small-Cell Lung/drug therapy , ErbB Receptors/genetics , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mutation , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/genetics , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Retinal Dehydrogenase/genetics , Signal Transduction , Tretinoin/pharmacology
8.
J Clin Invest ; 117(4): 889-901, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17380205

ABSTRACT

Duchenne muscular dystrophy (DMD) is a lethal X-linked disorder associated with dystrophin deficiency that results in chronic inflammation and severe skeletal muscle degeneration. In DMD mouse models and patients, we find that IkappaB kinase/NF-kappaB (IKK/NF-kappaB) signaling is persistently elevated in immune cells and regenerative muscle fibers. Ablation of 1 allele of the p65 subunit of NF-kappaB was sufficient to improve pathology in mdx mice, a model of DMD. In addition, conditional deletion of IKKbeta in mdx mice elucidated that NF-kappaB functions in activated macrophages to promote inflammation and muscle necrosis and in skeletal muscle fibers to limit regeneration through the inhibition of muscle progenitor cells. Furthermore, specific pharmacological inhibition of IKK resulted in improved pathology and muscle function in mdx mice. Collectively, these results underscore the critical role of NF-kappaB in the progression of muscular dystrophy and suggest the IKK/NF-kappaB signaling pathway as a potential therapeutic target for DMD.


Subject(s)
I-kappa B Kinase/metabolism , Macrophages/physiology , Muscle Fibers, Skeletal/physiology , Muscular Dystrophy, Duchenne/physiopathology , NF-kappa B/physiology , Signal Transduction/physiology , Animals , Disease Models, Animal , Disease Progression , Gene Deletion , Humans , Mice , Mice, Knockout , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/therapy , Transcription Factor RelA/genetics
9.
Mol Cell Biol ; 27(12): 4374-87, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17438126

ABSTRACT

NF-kappaB signaling is implicated as an important regulator of skeletal muscle homeostasis, but the mechanisms by which this transcription factor contributes to muscle maturation and turnover remain unclear. To gain insight into these mechanisms, gene expression profiling was examined in C2C12 myoblasts devoid of NF-kappaB activity. Interestingly, even in proliferating myoblasts, the absence of NF-kappaB caused the pronounced induction of several myofibrillar genes, suggesting that NF-kappaB functions as a negative regulator of late-stage muscle differentiation. Although several myofibrillar promoters contain predicted NF-kappaB binding sites, functional analysis using the troponin-I2 gene as a model revealed that NF-kappaB-mediated repression does not occur through direct DNA binding. In the search for an indirect mediator, the transcriptional repressor YinYang1 (YY1) was identified. While inducers of NF-kappaB stimulated YY1 expression in multiple cell types, genetic ablation of the RelA/p65 subunit of NF-kappaB in both cultured cells and adult skeletal muscle correlated with reduced YY1 transcripts and protein. NF-kappaB regulation of YY1 occurred at the transcriptional level, mediated by direct binding of the p50/p65 heterodimer complex to the YY1 promoter. Furthermore, YY1 was found associated with multiple myofibrillar promoters in C2C12 myoblasts containing NF-kappaB activity. Based on these results, we propose that NF-kappaB regulation of YY1 and transcriptional silencing of myofibrillar genes represent a new mechanism by which NF-kappaB functions in myoblasts to modulate skeletal muscle differentiation.


Subject(s)
Gene Expression Regulation, Developmental , Muscle Development/physiology , Myofibrils/genetics , NF-kappa B/metabolism , YY1 Transcription Factor/metabolism , Animals , Cell Culture Techniques , Cell Differentiation , Cell Line , Cell Line, Tumor , Cells, Cultured , Chromatin Immunoprecipitation , Gene Expression Profiling , Gene Silencing , Genes, Reporter , Humans , Immunohistochemistry , Luciferases/metabolism , Mice , Models, Biological , Myoblasts/cytology , Myoblasts/metabolism , NF-kappa B/genetics , Plasmids , Transfection , YY1 Transcription Factor/genetics
10.
Proc Natl Acad Sci U S A ; 104(49): 19506-11, 2007 Dec 04.
Article in English | MEDLINE | ID: mdl-18048329

ABSTRACT

The establishment of distant metastases depends on the capacity of small numbers of cancer cells to regenerate a tumor after entering a target tissue. The mechanisms that confer this capacity remain to be defined. Here we identify a role for the transcriptional inhibitors of differentiation Id1 and Id3 as selective mediators of lung metastatic colonization in the triple negative [TN, i.e., lacking expression of estrogen receptor and progesterone receptor, and lacking Her2 (human epidermal growth factor receptor 2) amplification] subgroup of human breast cancer. Although broad expression of Id1 has recently been documented in tumors of the rare metaplastic subtype, here we report that rare Id1-expressing cells are also present in the more common TN subset of human breast tumors but not in other subtypes. We also provide evidence that Id1 expression is enriched in clinically obtained hormone receptor negative lung metastases. Functional studies demonstrate that Id1 and its closely related family member Id3 are required for tumor initiating functions, both in the context of primary tumor formation and during metastatic colonization of the lung microenvironment. In vivo characterization of lung metastatic progression reveals that Id1 and Id3 facilitate sustained proliferation during the early stages of metastatic colonization, subsequent to extravasation into the lung parenchyma. These results shed light on the proliferative mechanisms that initiate metastatic colonization, and they implicate Id1 and Id3 as mediators of this malignant function in the TN subgroup of breast cancers.


Subject(s)
Breast Neoplasms/pathology , Inhibitor of Differentiation Protein 1/physiology , Inhibitor of Differentiation Proteins/physiology , Lung Neoplasms/genetics , Lung Neoplasms/secondary , Neoplasm Proteins/physiology , Animals , Cell Proliferation , Female , Humans , Inhibitor of Differentiation Protein 1/analysis , Inhibitor of Differentiation Protein 1/genetics , Inhibitor of Differentiation Proteins/analysis , Inhibitor of Differentiation Proteins/genetics , Lung Neoplasms/pathology , Mice , Mice, Transgenic , Neoplasm Proteins/analysis , Neoplasm Proteins/genetics , Transplantation, Heterologous
11.
Article in English | MEDLINE | ID: mdl-31615873

ABSTRACT

Metastasis arises when cancer cells disseminate from their site of origin and invade distant organs. While cancer cells rarely colonize muscle, they often induce a debilitating muscle-wasting condition known as cachexia that compromises feeding, breathing, and cardiac function in metastatic cancer patients. In fact, nearly 80% of metastatic cancer patients experience a spectrum of muscle-wasting states, which deteriorates the quality of life and overall survival of cancer patients. Muscle wasting in cancer results from increased muscle catabolism induced by circulating tumor factors and a systemic metabolic dysfunction. In addition, muscle loss can be exacerbated by the exposure to antineoplastic therapies and the process of aging. With no approved therapies to alleviate cachexia, muscle health, therefore, becomes a key determinant of prognosis, treatment response, and survival in metastatic cancer patients. This review will discuss the current understanding of cancer-associated cachexia and highlight promising therapeutic strategies to treat muscle wasting in the context of metastatic cancers.


Subject(s)
Cachexia/physiopathology , Muscular Atrophy/physiopathology , Neoplasm Metastasis/pathology , Aging , Antineoplastic Agents/adverse effects , Cachexia/etiology , Humans , Muscle, Skeletal/metabolism , Muscular Atrophy/etiology , Neoplasm Metastasis/drug therapy , Quality of Life
12.
Nat Rev Cancer ; 20(5): 274-284, 2020 05.
Article in English | MEDLINE | ID: mdl-32235902

ABSTRACT

Tumours reprogram host physiology, metabolism and immune responses during cancer progression. The release of soluble factors, exosomes and metabolites from tumours leads to systemic changes in distant organs, where cancer cells metastasize and grow. These tumour-derived circulating factors also profoundly impact tissues that are rarely inhabited by metastatic cancer cells such as skeletal muscle and adipose tissue. In fact, the majority of patients with metastatic cancer develop a debilitating muscle-wasting syndrome, known as cachexia, that is associated with decreased tolerance to antineoplastic therapy, poor prognosis and accelerated death, with no approved treatments. In this Perspective, we discuss the development of cachexia in the context of metastatic progression. We briefly discuss how circulating factors either directly or indirectly promote cachexia development and examine how signals from the metastatic process can trigger and amplify this process. Finally, we highlight promising therapeutic opportunities for targeting cachexia in the context of metastatic cancers.


Subject(s)
Cachexia/etiology , Neoplasms/complications , Animals , Cachexia/pathology , Disease Progression , Humans , Neoplasm Metastasis , Neoplasms/pathology
13.
Cancer Med ; 9(18): 6766-6775, 2020 09.
Article in English | MEDLINE | ID: mdl-32730698

ABSTRACT

Nearly 80% of advanced cancer patients are afflicted with cachexia, a debilitating syndrome characterized by extensive loss of muscle mass and function. Cachectic cancer patients have a reduced tolerance to antineoplastic therapies and often succumb to premature death from the wasting of respiratory and cardiac muscles. Since there are no available treatments for cachexia, it is imperative to understand the mechanisms that drive cachexia in order to devise effective strategies to treat it. Although 25% of metastatic breast cancer patients develop symptoms of muscle wasting, mechanistic studies of breast cancer cachexia have been hampered by a lack of experimental models. Using tumor cells deficient for BARD1, a subunit of the BRCA1/BARD1 tumor suppressor complex, we have developed a new orthotopic model of triple-negative breast cancer that spontaneously metastasizes to the lung and leads to systemic muscle deterioration. We show that expression of the metal-ion transporter, Zip14, is markedly upregulated in cachectic muscles from these mice and is associated with elevated intramuscular zinc and iron levels. Aberrant Zip14 expression and altered metal-ion homeostasis could therefore represent an underlying mechanism of cachexia development in human patients with triple-negative breast cancer. Our study provides a unique model for studying breast cancer cachexia and identifies a potential therapeutic target for its treatment.


Subject(s)
Cachexia/metabolism , Cation Transport Proteins/metabolism , Lung Neoplasms/metabolism , Muscle, Skeletal/metabolism , Triple Negative Breast Neoplasms/metabolism , Tumor Suppressor Proteins/deficiency , Ubiquitin-Protein Ligases/deficiency , Animals , BRCA1 Protein/metabolism , Cachexia/genetics , Cachexia/pathology , Cation Transport Proteins/genetics , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Lung Neoplasms/secondary , Mice , Muscle, Skeletal/pathology , Norisoprenoids/metabolism , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Tumor Suppressor Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Up-Regulation , Zinc/metabolism
14.
Cancers (Basel) ; 12(1)2019 Dec 18.
Article in English | MEDLINE | ID: mdl-31861290

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a lethal cancer type in which the mortality rate approaches the incidence rate. More than 85% of PDAC patients experience a profound loss of muscle mass and function, known as cachexia. PDAC patients with this condition suffer from decreased tolerance to anti-cancer therapies and often succumb to premature death due to respiratory and cardiac muscle wasting. Yet, there are no approved therapies available to alleviate cachexia. We previously found that upregulation of the metal ion transporter, Zip14, and altered zinc homeostasis are critical mediators of cachexia in metastatic colon, lung, and breast cancer models. Here, we show that a similar mechanism is likely driving the development of cachexia in PDAC. In two independent experimental metastasis models generated from the murine PDAC cell lines, Pan02 and FC1242, we observed aberrant Zip14 expression and increased zinc ion levels in cachectic muscles. Moreover, in advanced PDAC patients, high levels of ZIP14 in muscles correlated with the presence of cachexia. These studies underscore the importance of altered ZIP14 function in PDAC-associated cachexia development and highlight a potential therapeutic opportunity for improving the quality of life and prolonging survival in PDAC patients.

15.
Clin Cancer Res ; 13(5): 1356-61, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17332276

ABSTRACT

Cachexia is a life-threatening consequence of cancer that diminishes both quality of life and survival. It is a syndrome that is characterized by extreme weight loss resulting mainly from the depletion of skeletal muscle. Research from the past decades investigating the mechanisms of tumor-induced muscle wasting has identified several key cachectic factors that act through the ubiquitin-dependent proteasome system. Signaling pathways that mediate the effects of these cachectic factors have also subsequently emerged. Here, we review some of these pathways specific to myostatin, nuclear factor kappaB, and the newly elucidated dystrophin glycoprotein complex. Although these molecules are likely to employ distinct modes of action, results suggest that they nevertheless maintain a link to the proteasome pathway. Therefore, although the proteasome remains a preferred choice for therapy, the continually emerging upstream signaling molecules serve as additional promising therapeutic targets for the treatment of tumor-induced muscle wasting.


Subject(s)
Cachexia/etiology , Cachexia/physiopathology , Neoplasms/complications , Proteasome Endopeptidase Complex/physiology , Signal Transduction/physiology , Animals , Humans
16.
Eur J Transl Myol ; 28(2): 7590, 2018 Apr 24.
Article in English | MEDLINE | ID: mdl-29991992

ABSTRACT

A compounding feature of greater than 50% of all cancers is the high incidence of the cachexia syndrome, a complex metabolic disorder characterized by extreme weight loss due mainly to the gross depletion of skeletal muscle tissue. Although studies into the cause of cancer cachexia has spanned over multiple decades, little is known about the effects of various cancer treatments themselves on cachexia. For example, chemotherapy agents induce side effects such as nausea and anorexia, but these symptoms do not fully account for the changes seen with cancer cachexia. In this study we examine the effects of chemotherapeutic compounds, specifically, cisplatin in the colon-26 adenocarcinoma model of cancer cachexia. We find that although cisplatin is able to reduce tumor burden as expected, muscle wasting in mice nevertheless persists. Strikingly, cisplatin alone was seen to regulate muscle atrophy, which was independent of the commonly implicated ubiquitin proteasome system. Finally, we show that cisplatin is able to induce NF-κB activity in both mouse muscles and myotube cultures, suggesting that an additional side effect of cancer treatment is the regulation of muscle wasting that may be mediated through activation of the NF-κB signaling pathway.

17.
Nat Med ; 24(6): 770-781, 2018 06.
Article in English | MEDLINE | ID: mdl-29875463

ABSTRACT

Patients with metastatic cancer experience a severe loss of skeletal muscle mass and function known as cachexia. Cachexia is associated with poor prognosis and accelerated death in patients with cancer, yet its underlying mechanisms remain poorly understood. Here, we identify the metal-ion transporter ZRT- and IRT-like protein 14 (ZIP14) as a critical mediator of cancer-induced cachexia. ZIP14 is upregulated in cachectic muscles of mice and in patients with metastatic cancer and can be induced by TNF-α and TGF-ß cytokines. Strikingly, germline ablation or muscle-specific depletion of Zip14 markedly reduces muscle atrophy in metastatic cancer models. We find that ZIP14-mediated zinc uptake in muscle progenitor cells represses the expression of MyoD and Mef2c and blocks muscle-cell differentiation. Importantly, ZIP14-mediated zinc accumulation in differentiated muscle cells induces myosin heavy chain loss. These results highlight a previously unrecognized role for altered zinc homeostasis in metastatic cancer-induced muscle wasting and implicate ZIP14 as a therapeutic target for its treatment.


Subject(s)
Cachexia/metabolism , Cachexia/pathology , Cation Transport Proteins/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Neoplasms/metabolism , Neoplasms/pathology , Up-Regulation , Animals , Cell Differentiation , Cell Line , Cytokines/metabolism , Disease Models, Animal , Humans , Mice, Inbred C57BL , Myosin Heavy Chains/metabolism , Neoplasm Metastasis , Transforming Growth Factor beta/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Zinc/metabolism
18.
J Clin Invest ; 114(3): 370-8, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15286803

ABSTRACT

Cachexia is a syndrome characterized by wasting of skeletal muscle and contributes to nearly one-third of all cancer deaths. Cytokines and tumor factors mediate wasting by suppressing muscle gene products, but exactly which products are targeted by these cachectic factors is not well understood. Because of their functional relevance to muscle architecture, such targets are presumed to represent myofibrillar proteins, but whether these proteins are regulated in a general or a selective manner is also unclear. Here we demonstrate, using in vitro and in vivo models of muscle wasting, that cachectic factors are remarkably selective in targeting myosin heavy chain. In myotubes and mouse muscles, TNF-alpha plus IFN-gamma strongly reduced myosin expression through an RNA-dependent mechanism. Likewise, colon-26 tumors in mice caused the selective reduction of this myofibrillar protein, and this reduction correlated with wasting. Under these conditions, however, loss of myosin was associated with the ubiquitin-dependent proteasome pathway, which suggests that mechanisms used to regulate the expression of muscle proteins may be cachectic factor specific. These results shed new light on cancer cachexia by revealing that wasting does not result from a general downregulation of muscle proteins but rather is highly selective as to which proteins are targeted during the wasting state.


Subject(s)
Cachexia/metabolism , Colonic Neoplasms/complications , Gene Expression Regulation, Neoplastic , Muscle, Skeletal/metabolism , MyoD Protein/metabolism , Myosin Heavy Chains/metabolism , Animals , CHO Cells , Cell Line, Tumor , Colonic Neoplasms/metabolism , Cricetinae , Cricetulus , Cysteine Endopeptidases/metabolism , Disease Models, Animal , Down-Regulation , Drug Synergism , Gene Targeting , Interferon-gamma/metabolism , Interferon-gamma/pharmacology , Mice , Mice, Inbred Strains , Mice, Nude , Multienzyme Complexes/metabolism , Muscle, Skeletal/drug effects , MyoD Protein/drug effects , MyoD Protein/genetics , Myosin Heavy Chains/drug effects , Myosin Heavy Chains/genetics , Proteasome Endopeptidase Complex , Sensitivity and Specificity , Transcription, Genetic/drug effects , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Ubiquitins/metabolism
19.
Dalton Trans ; 46(33): 10963-10985, 2017 Aug 22.
Article in English | MEDLINE | ID: mdl-28766668

ABSTRACT

Refluxing [VIVO(ß-diketonate)2], namely [VIVO(acetylacetonate)2] and [VIVO(benzoylacetonate)2], separately with an equivalent or excess amount of 2-aminobenzoylhydrazide (ah) in laboratory grade (LG) CH3OH in aerobic conditions afforded non-oxidovanadium(iv) and oxidovanadium(v) complexes of the type [VIV(L1)2] (1), [VVO(L1)(OCH3)]2 (3) and [VIV(L2)2] (2), and [VVO(L2)(OCH3)] (4), respectively. (L1)2- and (L2)2- represent the dianionic forms of 2-aminobenzoylhydrazone of acetylacetone (H2L1) and benzoylacetone (H2L2), respectively, (general abbreviation, H2L), which was formed by the in situ condensation of ah with the respective coordinated [ß-diketonate] in medium-to-good yield. The yield of different resulting products was dependent upon the ratio of ah to [VIVO(ß-diketonate)2]. For example, the yield of 1 and 2 complexes increased significantly associated with a decrease in the amount of 3 and 4 with an increase in the molar ratio of ah. Upon replacing CH3OH by a non-hydroxylic solvent, LG CHCl3, the above reaction yielded only oxidovanadium(v) complexes of the type [VVO(L1)(OH)]2 (5), [VVO(L2)(OH)] (6) and [VO3(L)2] (7, 8) whereas, upon replacing CHCl3 by another non-hydroxylic solvent, namely LG CH3CN, only the respective [VO3(L)2] (7, 8) complex was isolated in 72-78% yield. However, upon performing the above reactions in the absence of air using dry CH3OH or dry CHCl3, only the respective [VIV(L)2] complex was obtained, suggesting that aerial oxygen was the oxidising agent and the type of pentavalent product formed was dependent upon the nature of solvent used. Complexes 3 and 4 were converted, respectively, to 7 and 8 on refluxing in LG CHCl3via the respective unstable complex 5 and 6. The DFT calculated change in internal energy (ΔE) for the reactions 2[VVO(L2)(OCH3)] + 2H2O → 2[VVO(L2)(OH)] + 2CH3OH and 2[VVO(L2)(OH)] → [VO3(L2)2] + H2O was, respectively, +3.61 and -7.42 kcal mol-1, suggesting that the [VVO(L2)(OH)] species was unstable and readily transformed to the stable [VO3(L2)2] complex. Upon one-electron reduction at an appropriate potential, each of 7 and 8 generated mixed-valence [(L)VVO-(µ-O)-OVIV(L)]- species, which showed valence-delocalisation at room temperature and localisation at 77 K. Some of the complexes showed a wide range of toxicity in a dose-dependent manner against lung cancer cells comparable with that observed with cis-platin.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Hydrazines/chemistry , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacology , Solvents/chemistry , Vanadium/chemistry , Aerobiosis , Anaerobiosis , Cell Line, Tumor , Humans
20.
Cell Res ; 26(3): 273-4, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26823207

ABSTRACT

It is becoming increasingly clear that leukocytes dynamically regulate cancer progression and metastasis, and among leukocytes, granulocytic cells abundantly accumulate in metastatic organs; however, their function in metastasis remains controversial. In a recent report in Nature, Wculek and Malanchi clarify the role of mature neutrophils as mediators of metastatic initiation and provide a targeted approach to prevent the pro-metastatic activity of neutrophils in breast cancer models.


Subject(s)
Breast Neoplasms/pathology , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Neoplasm Metastasis/pathology , Neutrophils/metabolism , Animals , Female
SELECTION OF CITATIONS
SEARCH DETAIL