Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Joseph H. McCarty is active.

Publication


Featured researches published by Joseph H. McCarty.


Nature Medicine | 2013

Targeting of αv integrin identifies a core molecular pathway that regulates fibrosis in several organs.

Neil C. Henderson; Thomas D. Arnold; Yoshio Katamura; Marilyn M. Giacomini; Juan Rodríguez; Joseph H. McCarty; Antonella Pellicoro; Elisabeth Raschperger; Christer Betsholtz; Peter Ruminski; David W. Griggs; Michael J. Prinsen; Jacquelyn J. Maher; John P. Iredale; Adam Lacy-Hulbert; Ralf H. Adams; Dean Sheppard

Myofibroblasts are the major source of extracellular matrix components that accumulate during tissue fibrosis, and hepatic stellate cells (HSCs) are believed to be the major source of myofibroblasts in the liver. To date, robust systems to genetically manipulate these cells have not been developed. We report that Cre under control of the promoter of Pdgfrb (Pdgfrb-Cre) inactivates loxP-flanked genes in mouse HSCs with high efficiency. We used this system to delete the gene encoding αv integrin subunit because various αv-containing integrins have been suggested as central mediators of fibrosis in multiple organs. Such depletion protected mice from carbon tetrachloride–induced hepatic fibrosis, whereas global loss of β3, β5 or β6 integrins or conditional loss of β8 integrins in HSCs did not. We also found that Pdgfrb-Cre effectively targeted myofibroblasts in multiple organs, and depletion of the αv integrin subunit using this system was protective in other models of organ fibrosis, including pulmonary and renal fibrosis. Pharmacological blockade of αv-containing integrins by a small molecule (CWHM 12) attenuated both liver and lung fibrosis, including in a therapeutic manner. These data identify a core pathway that regulates fibrosis and suggest that pharmacological targeting of all αv integrins may have clinical utility in the treatment of patients with a broad range of fibrotic diseases.


Nature | 2015

Microenvironment-induced PTEN loss by exosomal microRNA primes brain metastasis outgrowth

Lin Zhang; Siyuan Zhang; Jun Yao; Frank J. Lowery; Qingling Zhang; Wen Chien Huang; Ping Li; Min Li; Xiao Wang; Chenyu Zhang; Hai Wang; Kenneth Ellis; Mujeeburahiman Cheerathodi; Joseph H. McCarty; Diane Palmieri; Jodi M. Saunus; Sunil R. Lakhani; Suyun Huang; Aysegul A. Sahin; Kenneth D. Aldape; Patricia S. Steeg; Dihua Yu

The development of life-threatening cancer metastases at distant organs requires disseminated tumour cells’ adaptation to, and co-evolution with, the drastically different microenvironments of metastatic sites. Cancer cells of common origin manifest distinct gene expression patterns after metastasizing to different organs. Clearly, the dynamic interaction between metastatic tumour cells and extrinsic signals at individual metastatic organ sites critically effects the subsequent metastatic outgrowth. Yet, it is unclear when and how disseminated tumour cells acquire the essential traits from the microenvironment of metastatic organs that prime their subsequent outgrowth. Here we show that both human and mouse tumour cells with normal expression of PTEN, an important tumour suppressor, lose PTEN expression after dissemination to the brain, but not to other organs. The PTEN level in PTEN-loss brain metastatic tumour cells is restored after leaving the brain microenvironment. This brain microenvironment-dependent, reversible PTEN messenger RNA and protein downregulation is epigenetically regulated by microRNAs from brain astrocytes. Mechanistically, astrocyte-derived exosomes mediate an intercellular transfer of PTEN-targeting microRNAs to metastatic tumour cells, while astrocyte-specific depletion of PTEN-targeting microRNAs or blockade of astrocyte exosome secretion rescues the PTEN loss and suppresses brain metastasis in vivo. Furthermore, this adaptive PTEN loss in brain metastatic tumour cells leads to an increased secretion of the chemokine CCL2, which recruits IBA1-expressing myeloid cells that reciprocally enhance the outgrowth of brain metastatic tumour cells via enhanced proliferation and reduced apoptosis. Our findings demonstrate a remarkable plasticity of PTEN expression in metastatic tumour cells in response to different organ microenvironments, underpinning an essential role of co-evolution between the metastatic cells and their microenvironment during the adaptive metastatic outgrowth. Our findings signify the dynamic and reciprocal cross-talk between tumour cells and the metastatic niche; importantly, they provide new opportunities for effective anti-metastasis therapies, especially of consequence for brain metastasis patients.


Development | 2004

Selective ablation of αv integrins in the central nervous system leads to cerebral hemorrhage, seizures, axonal degeneration and premature death

Joseph H. McCarty; Adam Lacy-Hulbert; Alain Charest; Roderick T. Bronson; Denise Crowley; David E. Housman; John Savill; Jürgen Roes; Richard O. Hynes

Mouse embryos genetically null for all αv integrins develop intracerebral hemorrhage owing to defective interactions between blood vessels and brain parenchymal cells. Here, we have used conditional knockout technology to address whether the cerebral hemorrhage is due to primary defects in vascular or neural cell types. We show that ablating αv expression in the vascular endothelium has no detectable effect on cerebral blood vessel development, whereas deletion of αv expression in central nervous system glial cells leads to embryonic and neonatal cerebral hemorrhage. Conditional deletion of αv integrin in both central nervous system glia and neurons also leads to cerebral hemorrhage, but additionally to severe neurological defects. Approximately 30% of these mutants develop seizures and die by 4 weeks of age. The remaining mutants survive for several months, but develop axonal deterioration in the spinal cord and cerebellum, leading to ataxia and loss of hindlimb coordination. Collectively, these data provide evidence that αv integrins on embryonic central nervous system neural cells, particularly glia, are necessary for proper cerebral blood vessel development, and also reveal a novel function for αv integrins expressed on axons in the postnatal central nervous system.


Molecular and Cellular Biology | 2002

Defective Associations between Blood Vessels and Brain Parenchyma Lead to Cerebral Hemorrhage in Mice Lacking αv Integrins

Joseph H. McCarty; Rita A. Monahan-Earley; Lawrence F. Brown; Markus Keller; Holger Gerhardt; Kristofer Rubin; Moshe Shani; Harold F. Dvorak; Hartwig Wolburg; Bernhard L. Bader; Ann M. Dvorak; Richard O. Hynes

ABSTRACT Mouse embryos genetically null for the αv integrin subunit develop intracerebral hemorrhages at midgestation and die shortly after birth. A key question is whether the hemorrhage arises from primary defects in vascular endothelial cells or pericytes or from other causes. We have previously reported normal initiation of cerebral vessels comprising branched tubes of endothelial cells. Here we show that the onset of hemorrhage is not due to defects in pericyte recruitment. Additionally, most αv-null vessels display ultrastructurally normal endothelium-pericyte associations and normal interendothelial cell junctions. Thus, endothelial cells and pericytes appear to establish their normal relationships in cerebral microvessels. However, by both light and electron microscopy, we detected defective associations between cerebral microvessels and the surrounding brain parenchyma, composed of neuroepithelial cells, glia, and neuronal precursors. These data suggest a novel role for αv integrins in the association between cerebral microvessels and central nervous system parenchymal cells.


Cancer Research | 2011

Glioblastoma angiogenesis and tumor cell invasiveness are differentially regulated by β8 integrin

Jeremy H. Tchaicha; Steve B. Reyes; Jaekyung Shin; Mohammad G. Hossain; Frederick F. Lang; Joseph H. McCarty

Glioblastoma multiforme (GBM) is a highly invasive brain tumor that develops florid microvascular proliferation and hemorrhage. However, mechanisms that favor invasion versus angiogenesis in this setting remain largely uncharacterized. Here, we show that integrin β8 is an essential regulator of both GBM-induced angiogenesis and tumor cell invasiveness. Highly angiogenic and poorly invasive tumors expressed low levels of β8 integrin, whereas highly invasive tumors with limited neovascularization expressed high levels of β8 integrin. Manipulating β8 integrin protein levels altered the angiogenic and invasive growth properties of GBMs, in part, reflected by a diminished activation of latent TGFβs, which are extracellular matrix protein ligands for β8 integrin. Taken together, these results establish a role for β8 integrin in differential control of angiogenesis versus tumor cell invasion in GBM. Our findings suggest that inhibiting β8 integrin or TGFβ signaling may diminish tumor cell invasiveness during malignant progression and following antivascular therapies.


Laboratory Investigation | 2011

TGF-β signaling in endothelial cells, but not neuroepithelial cells, is essential for cerebral vascular development

Ha Long Nguyen; Young Jae Lee; Jaekyung Shin; Eun-Ji Lee; Sung Ok Park; Joseph H. McCarty; S. Paul Oh

The various organs of the body harbor blood vessel networks that display unique structural and functional features; however, the mechanisms that control organ-specific vascular development and physiology remain mostly unknown. In the developing mouse brain, αvβ8 integrin-mediated TGF-β activation and signaling is essential for normal blood vessel growth and sprouting. Whether integrins activate TGF-β signaling pathways in vascular endothelial cells (ECs), neural cells, or both, has yet to be determined. Here, we have generated and characterized mice in which TGF-β receptors are specifically deleted in neuroepithelial cells via Nestin-Cre, or in ECs via a novel Cre transgenic strain (Alk1GFPCre) in which Cre is expressed under control of the endogenous activin receptor-like kinase 1 (Alk1) promoter. We report that deletion of Tgfbr2 in the neuroepithelium does not impact brain vascular development. In contrast, selective deletion of the Tgfbr2 or Alk5 genes in ECs result in embryonic lethality because of brain-specific vascular pathologies, including blood vessel morphogenesis and intracerebral hemorrhage. These data reveal for the first time that αvβ8 integrin-activated TGF-βs regulate angiogenesis in the developing brain via paracrine signaling to ECs.


Development | 2011

The astrocyte-expressed integrin αvβ8 governs blood vessel sprouting in the developing retina.

Shinya Hirota; Qian Liu; Hye Shin Lee; Mohammad G. Hossain; Adam Lacy-Hulbert; Joseph H. McCarty

The mouse retina is vascularized after birth when angiogenic blood vessels grow and sprout along a pre-formed latticework of astrocytes. How astrocyte-derived cues control patterns of blood vessel growth and sprouting, however, remains enigmatic. Here, we have used molecular genetic strategies in mice to demonstrate that αvβ8 integrin expressed in astrocytes is essential for neovascularization of the developing retina. Selective ablation of αv or β8 integrin gene expression in astrocytes leads to impaired blood vessel sprouting and intraretinal hemorrhage, particularly during formation of the secondary vascular plexus. These pathologies correlate, in part, with diminished αvβ8 integrin-mediated activation of extracellular matrix-bound latent transforming growth factor βs (TGFβs) and defective TGFβ signaling in vascular endothelial cells, but not astrocytes. Collectively, our data demonstrate that αvβ8 integrin is a component of a paracrine signaling axis that links astrocytes to blood vessels and is essential for proper regulation of retinal angiogenesis.


Molecular Biology of the Cell | 2013

αvβ8 integrin interacts with RhoGDI1 to regulate Rac1 and Cdc42 activation and drive glioblastoma cell invasion

Steve B. Reyes; Anjana S. Narayanan; Hye Shin Lee; Jeremy H. Tchaicha; Kenneth D. Aldape; Frederick F. Lang; Kimberly F. Tolias; Joseph H. McCarty

Experiments with human cancer glioblastoma multiforme cell lines, primary patient samples, and preclinical mouse models are performed to show that αvβ8 integrin and RhoGDI1 are components of a signaling axis that drives brain tumor cell invasion via regulation of Rho GTPase activation.


Molecular and Cellular Biology | 2005

Loss of the Putative Tumor Suppressor Band 4.1B/Dal1 Gene Is Dispensable for Normal Development and Does Not Predispose to Cancer

Chunling Yi; Joseph H. McCarty; Scott Troutman; Matthew S. Eckman; Roderick T. Bronson; Joseph L. Kissil

ABSTRACT The band 4.1 proteins are cytoskeletal proteins, harboring a conserved FERM domain highly homologous to the N-terminal FERM domain of ezrin, radixin, moesin, and merlin. Recently, a truncated form of the 4.1B protein, termed Dal-1, was identified in a screen as down regulated in adenocarcinoma of the lung and was mapped to chromosome 18p11.3, which is lost in 38% of primary non-small cell lung carcinoma tumors. Analysis of several meningiomas has shown that Dal-1 expression was lost in 76% of the tumors. To further elucidate the function of the 4.1B/Dal-1 gene in development and tumorigenesis we generated mice deficient for this allele. The 4.1B/Dal-1 null mice develop normally and are fertile. Rates of cellular proliferation and apoptosis in brain, mammary, and lung tissues from the 4.1B/Dal-1 null mice were indistinguishable from those seen with wild-type mice. Aging studies indicate that these mice do not have a propensity to develop tumors. Analysis of fibroblasts from these mice demonstrated that the growth characteristics and kinetics of these cells were not different from those of cells from the wild-type mice. These findings indicate that the 4.1B gene is not required for normal development and that 4.1B/Dal-1 does not function as a tumor suppressor gene.


Oncogene | 2010

A mosaic mouse model of astrocytoma identifies alphavbeta8 integrin as a negative regulator of tumor angiogenesis.

Jeremy H. Tchaicha; Aaron K. Mobley; Mohammad G. Hossain; Kenneth D. Aldape; Joseph H. McCarty

Angiogenesis involves a complex set of cell–cell and cell–extracellular matrix (ECM) interactions that coordinately promote and inhibit blood vessel growth and sprouting. Although many factors that promote angiogenesis have been characterized, the identities and mechanisms of action of endogenous inhibitors of angiogenesis remain unclear. Furthermore, little is known about how cancer cells selectively circumvent the actions of these inhibitors to promote pathological angiogenesis, a requisite event for tumor progression. Using mosaic mouse models of the malignant brain cancer, astrocytoma, we report that tumor cells induce pathological angiogenesis by suppressing expression of the ECM protein receptor αvβ8 integrin. Diminished integrin expression in astrocytoma cells leads to reduced activation of latent TGFβs, resulting in impaired TGFβ receptor signaling in tumor-associated endothelial cells. These data reveal that astrocytoma cells manipulate their angiogenic balance by selectively suppressing αvβ8 integrin expression and function. Finally, these results show that an adhesion and signaling axis normally involved in developmental brain angiogenesis is pathologically exploited in adult brain tumors.

Collaboration


Dive into the Joseph H. McCarty's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mohammad G. Hossain

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Aaron K. Mobley

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Paola A. Guerrero

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Richard O. Hynes

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Kenneth D. Aldape

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Frederick F. Lang

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

John E. Morales

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Hye Shin Lee

Seoul National University

View shared research outputs
Researchain Logo
Decentralizing Knowledge